Next Article in Journal
BrWRKY65, a WRKY Transcription Factor, Is Involved in Regulating Three Leaf Senescence-Associated Genes in Chinese Flowering Cabbage
Next Article in Special Issue
Deconstructing Signaling Pathways in Cancer for Optimizing Cancer Combination Therapies
Previous Article in Journal
Activation of Magnesium Lignosulfonate and Kraft Lignin: Influence on the Properties of Phenolic Resin-Based Composites for Potential Applications in Abrasive Materials
Previous Article in Special Issue
Down’s Syndrome and Triple Negative Breast Cancer: A Rare Occurrence of Distinctive Clinical Relationship
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alteration of SHP-1/p-STAT3 Signaling: A Potential Target for Anticancer Therapy

1
Comprehensive Breast Health Center, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taiwan
2
Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taiwan
3
Institute of Biopharmaceutical Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei 112, Taiwan
4
Faculty of Pharmacy, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei 112, Taiwan
5
School of Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Taipei 112, Taiwan
6
Department of Medical Research, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
7
National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(6), 1234; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061234
Submission received: 2 May 2017 / Revised: 1 June 2017 / Accepted: 5 June 2017 / Published: 8 June 2017
(This article belongs to the Special Issue Alterations to Signalling Pathways in Cancer Cells)

Abstract

:
The Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 1 (SHP-1), a non-receptor protein tyrosine phosphatase, has been reported as a negative regulator of phosphorylated signal transducer and activator of transcription 3 (STAT3) and linked to tumor development. In this present review, we will discuss the importance and function of SHP-1/p-STAT3 signaling in nonmalignant conditions as well as malignancies, its cross-talk with other pathways, the current clinical development and the potential role of inhibitors of this pathway in anticancer therapy and clinical relevance of SHP-1/p-STAT3 in cancers. Lastly, we will summarize and highlight work involving novel drugs/compounds targeting SHP-1/p-STAT3 signaling and combined strategies that were/are discovered in our and our colleagues’ laboratories.

Graphical Abstract

1. Introduction

Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor which functions mainly through dimerization upon phosphorylation at tyrosine residues and translocation to cell nuclei [1,2]. STAT3 signaling is constitutively activated in various malignant human cancers and participating in multiple cellular progress as well as tumorigenesis [3,4]. Targeting STAT3 in cancer treatment has shown therapeutic benefits in both preclinical and clinical studies [5,6,7]. Common strategies for inhibiting STAT3 signaling includes inhibition of upstream receptors (Janus associated kinase (JAKs), interleukin-6 receptor), interfering STAT3 domain (block dimerization), impedance of STAT3-DNA binding, and inhibition of STAT3 transcription (by anti-sense oligonucleotides) and there have been corresponding investigational drugs/compounds to each strategy in various developmental stages [4,8,9,10,11]. Currently, no STAT3 inhibitors for cancer therapy have yet been approved, regardless of the mechanisms of STAT3 inhibition. There are several literatures that have nicely reviewed the progress of STAT3 inhibitors [10,11,12,13,14]. While many studies demonstrated that over-activation of Janus associated kinase (JAK) or growth factor receptor-associated tyrosine kinase (Src) contribute to the hyper-phosphorylation of STAT3 [15,16], phosphorylation of STAT3 is also tightly regulated by protein tyrosine phosphatases (PTPs) [17,18]. The Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 1 (SHP-1), a non-receptor SH2 domain-containing PTP, has been reported to dephosphorylate STAT3 at its tyrosine 705 (Tyr 705) residue directly [19]. Recently, abnormal SHP-1/p-STAT3 signaling pathway was identified in various human malignant tumors, including multiple myeloma [20], hepatocellular carcinoma [21], breast cancer [22] and triple-negative breast cancer [23]. Therefore, targeting p-STAT3 by enhancing the activities of SHP-1 may be a strategy for inhibiting p-STAT3 in cancers. In this review, we detailed the recent knowledge of SHP-1/STAT3 signaling in cancer progression and potent anti-cancer agents that can target this pathway.

2. SHP-1/p-STAT3 Pathway in Cancers

Oncogenic STAT3 hyper-activation has been observed in various malignant human tumors, including lung, breast, colon, liver, prostate, stomach, pancreas, kidney and brain cancers [3]. The major phosphorylation sites in STAT3 are Tyr 705 and Ser 727. STAT3 activation induces the various cellular signaling required for cell survival (e.g., Bcl-xl, Bcl2, c-Myc, Mcl-1 and survivin), proliferation (e.g., cyclin D1), migration and invasion (e.g., MMP-2, MMP-7, MMP-9, Rho and Rac), as well as angiogenesis (e.g., vascular endothelial growth factor (VEGF) [4,11,24,25]. Besides its normal function, STAT3 has been shown to involve in the tumor development and cell transformation. Pioneering work indicates that constitutive STAT3 activation is found in v-Src-transformed cell lines [26] and is sufficient to induce transformation of normal breast or prostate epithelial cell lines and immortalized fibroblast [27]. These findings indicate that abnormal STAT3 expression can cause permanent changes in gene expression program and lead to a malignant phenotype.
To prevent inappropriately sustained STAT3 activation, there are multiple intrinsic regulators that can timely suppress STAT3 pathway. SHP-1 is one of PTP members that can accomplish this important function [28]. SHP-1 has been shown to dephosphorylate JAK kinases [29,30,31,32] and STAT3 directly [19] to silence the JAK/STAT pathway. SHP-1 is composed of three domains including N-SH2 domain, C-SH2 domain and catalytic PTP domain [33]. SHP-1 forms an auto-inhibitory conformation from N-SH2 domain to catalytic domain and maintains its enzyme activity in the inactive state [33,34]. SHP-1 has been shown to function as a tumor suppressor to inhibit the tumor growth including breast cancer [35], pancreatic cancer [36] and prostate cancer [37]. Previous studies have reported that SHP-1 is highly expressed in normal lymphoid cells, but diminished or abolished in many types of cancer cell lines and tumor tissues [38,39]. Growing evidence has revealed that loss of SHP-1 leads to constitutive activation of STAT3 in hematologic malignancies, since ectopic expression of SHP-1 in leukemia cells substantially decreases the levels of activated STAT3 [40]. Interestingly, STAT3 promotes epigenetic SHP-1 gene silencing by forming complexes with DNA methyltransferase 1 (DNMT1) and histone deacetylase 1 (HDAC1) at the promoter region of SHP-1 [41]. Noteworthy, STAT3 activation usually accompanies high expression levels of DNMT1 to make an effective SHP-1 gene silencing [41]. Taken together, loss of SHP-1 and sustained activated STAT3 activation may cause an oncogenic feedforward loop to render the malignant cells more sensitive to a series of extra- and intracellular stimuli.
The central link of STAT3 with other key oncogenic pathways such as activated protein 1 (AP-1) signaling complex, nuclear factor NF-κB and Wnt/β-catenin signaling makes STAT3 an attractive target and a master regulator for a plethora of cellular functions. For example, STAT3 regulates a broad range of transcription factors, such as c-fos, whereas c-fos is a key member of AP-1 proteins, a key cell life and death regulator [42]. The combination of STAT3 and AP-1 activities have been shown to drive elevated MMP-1 expression and promote colorectal cancer (CRC) invasion [43]. The cross-regulation between the Wnt/β-catenin and NF-κB signaling plays an important role in a diverse array of genes and pathways responsible for chronic inflammation, immunity, development, and tumorigenesis [44]. Aberrant activation and interaction of STAT3 and Wnt/β-catenin occurs in malignancies [45,46], and the convergence of these two pathways could regulate cell survival and stemness [47,48,49]. Moreover, NF-κB and STAT3 are crucial for controlling the abilities to resist apoptosis-based tumor surveillance as well as regulating angiogenesis and invasiveness in preneoplastic and malignant cells [50]. Notably, NF-κB and STAT3 can cooperate to promote cancer development and progression [51], and also regulate distinct functions in surrounding non-tumorigenic cells [52]. These key transcription factors NF-κB and AP-1 were also strongly activated in the absence of SHP-1 [53,54,55]. In addition, SHP-1 negative-regulated β-catenin transcriptional function and intestinal epithelial cell proliferation [56]. These studies further supported the cross-talk networks between the SHP-1/p-STAT3 pathway and these oncogenic signal transduction cascades. In addition to investigating the molecular mechanisms of cancer progression, these interactions also offer new insight into developing anti-cancer agents [51]. Future studies may help to delineate the impact of targeting SHP-1/STAT3 on the network among these pathways.

3. SHP-1/STAT3 Pathway Is a Target in the Treatment of Human Malignancies

SHP-1-mediated STAT3 downregulation is an appealing anti-cancer strategy to induce apoptosis in cancer cells. Previously, sorafenib has been demonstrated that can induce apoptosis in cancer cells through a novel kinase inhibition-independent mechanism. Sorafenib is the first [57] and is still the only Food and Drug Administration (FDA)-approved targeted therapy for advanced hepatocellular carcinoma cells (HCC) in 2016. Sorafenib has been reported to induce cell growth arrest and apoptosis in variety cancers including medulloblastomas [58], pancreatic cancer [59], glioblastoma [60], neuroblastoma [61], acute myeloid leukemia (AML) [62] and hepatocellular carcinoma (HCC) cells [63]. Our group has identified that sorafenib targets STAT3 in a kinase-independent pathway [19] and further generated a series of sorafenib derivatives (SC compounds such as SC-1, SC-40, SC-43, SC-49, SC-60 and SC-78) which lack activities on kinases but effectively induce cell apoptosis in cancers [19,64,65]. Sorafenib is a multiple kinase inhibitor targeting Raf-1 and other tyrosine kinases (e.g., VEGFR2, VEGFR3, Flt-3, PDGFR, and FGFR-1) [66,67]. In our works, sorafenib, but not its derivatives SC-1 [68], SC-43 [68] and SC-60 [69], significantly decrease the activity of Raf-1 kinase as well as the phosphorylation of VEGFR2 and PDGFRβ. We also proved that sorafenib and its analogues SC-1 and SC-43 showed no obvious effects on the phosphorylation of STAT3 upstream regulator JAK1 or JAK2, but effectively decreased the p-STAT3 proteins [68]. Sorafenib increased the enzyme activity of SHP-1 by directly interacting and impairing the association between the N-SH2 domain and the catalytic protein tyrosine phosphatase domain of SHP-1 [70]. We found that the N-terminal SH2 domain is a critical docking site of sorafenib [70]. Sorafenib derivatives SC-40 and SC-43, two potent SHP-1 enhancers, were also docked in the same site [70]. We therefore hypothesized that the interaction of sorafenib (or its derivatives SC-43 and SC-60) and the N-SH2 domain might lead to a release of the D61 catalytic site and activation of SHP-1 activity. Currently, the hypothesized mechanism was supported by using ectopic expressing dN1 (deleted N-SH2) and D61A mutant SHP-1 in cholangiocarcinoma [71], HCC [72], CRC [73], and triple-negative breast cancer (TNBC) [69] cells. Compared to wild-type SHP-1-expressing cells, SC-43 [71,73] and SC-60 [69,72] exerted less p-STAT3 downregulation and apoptosis-promoting effects on these mutant SHP-1-expressing cells. Compared with sorafenib, SC-1 and SC-43 induced more potent apoptosis in association with downregulation of p-STAT3 and its downstream molecules (cyclin D1 and survivin) in breast cancer cell lines [68]. The same mechanism existed in SC-60, a dimer-based sorafenib derivative that successfully exhibits anti-cancer ability in HCC [72] and TNBC [69]. Besides sorafenib, regorafenib, which is an inhibitor of multiple protein kinases, also exerts anti-tumor ability by reactivating SHP-1 and inhibiting STAT3 signaling in metastatic CRC [73,74] and HCC [75]. Su et al. also found that regorafenib and its analogue SC-78 inhibit cell growth and metastasis through SHP-1/p-STAT3/VEGF-A axis in TNBC [23]. Dovitinib, another multiple kinase inhibitor, induces significant apoptosis in HCC cells and sorafenib-resistant cells [76] by SHP-1-dependent STAT3 inhibition. SHP-1 has also been demonstrated as a target of nintedanib, which is a triple angiokinase inhibitor. Nintedanib induces HCC cell apoptosis by relieving the autoinhibitory structure of SHP-1 in an angiokinase-independent pathway [77]. Capillarisin which is derived from Artemisia capillaris induces SHP-1 to downregulate the expression of Jak1/2 and STAT3, and STAT3-regulated genes that mediate cell proliferation, cell survival, invasion, and angiogenesis [78].
Apart from the above-mentioned drugs, there is a range of other anticancer compounds like Icariside II [79], betulinic acid [80], ergosterol peroxide [81], epigallocatechin-3-gallate [82], genipin [83], boswellic acid [84], ginkgolic acid C 17:1 [85], emodin [86], γ-Tocotrienol [87], honokiol [88], pectolinarigenin [89], and [90] zerumbone that modulates the expression or activity of SHP-1 to decrease the p-STAT3 proteins. Luteolin disrupts the interaction of HSP-90 and STAT3 and enhances SHP-1 to dephosphorylate STAT3 [91]. Hence, discovery and development of SHP-1 agonists may be a promising approach to combat tumor progression, drug resistance, and improve the survival rate as well as life quality of patients suffering from cancers. These findings suggest that SHP-1-mediated STAT3 downregulation is the potential target of anticancer drugs to inhibit cell growth and induce apoptosis in cancer cells. The regulation of SHP-1/p-STAT3 pathway and potent SHP-1 agonists are diagrammatically explained in Figure 1.

4. Selective Targeting SHP-1 Is Augmented by Combination Therapy with Chemotherapeutic Agents for STAT3 Signaling Blockade

Despite these promising results, SHP-1 agonist in combination with current approved chemotherapeutic agents may be synergistic to overcome toxicity, drug resistance or other side effects associated with high doses of single drugs. Sorafenib was the first and remains the only approved targeted therapy for advanced HCC in 2016. Comparison with single-agent sorafenib, sorafenib in combination with SC-43 had a synergistic effect on the increment of SHP-1 activity and the decrement of p-STAT3 in HCC cells [64]. In addition, combined sorafenib with SC-43 significantly promoted the apoptotic effect on sorafenib-resistant HCC cells [70]. Sorafenib and its derivative SC-49 also sensitized HCC cells to a novel anti-human death receptor 5 monoclonal antibody CS-1008-induced apoptosis through SHP-1-dependent STAT3 inhibition [92]. In radiation therapy for HCC, SC-59, which is another novel SHP-1 agonist, showed a better synergistic effect than sorafenib [93]. Combination therapy of sorafenib and SC-2001 (Mcl-1 inhibitor) inhibited STAT3 activation by RFX-dependent SHP-1 reactivation and defeated the sorafenib resistance in HCC and breast cancer cells [22,94,95]. Before the approval of sorafenib, the use of doxorubicin was common in advanced HCC treatment. Single-agent doxorubicin showed a 79% response rate in an initial phase II study [96]. However, subsequent studies exhibited limited efficacy and no obvious survival benefit [97,98,99,100,101]. Recently, potent SHP-1 agonists such as emodin [86], honokiol [88] and g-tocotrienol [87] effectively enhanced the apoptotic effect of doxorubicin and paclitaxel in HCC cells. γ-tocotrienol in combination with EGFR inhibitors (erlotinib or gefitinib) suppressed STAT3 and Akt signaling in murine mammary tumor cells [102]. Betulinic acid enhanced apoptosis induced by thalidomide and bortezomib in human multiple myeloma cells [80]. Dovitinib also acted as a novel radiosensitizer [103] and sensitized HCC cells to TNF-related apoptosis-inducing ligand (TRAIL) and tigatuzumab [104]. 5-azacytidine sensitized FLT3-ITD positive AML to lestaurtinib (CEP-701) by upregulating SHP-1 expression [105]. The combination of sorafenib and YC-1 (a soluble guanylyl cyclase activator) significantly displayed an anti-HCC effect compared with sorafenib or YC-1 used alone by modulating SHP-1/p-STAT3 pathway [106]. Plumbagin, a vitamin K3 analogue, has also been reported to inhibit STAT3 signaling by induction of SHP-1 and may have potential to sensitize STAT3-overexpressing cancers to thalidomide and bortezomib [107]. These studies provide the foundation for SHP-1/p-STAT3 modulators in conjunction with approved targeted agents in future cancer therapy.

5. Clinical Relevance of SHP-1/p-STAT3 in Cancers

Notwithstanding, the prognostic role of p-STAT3 on cancer patient outcome seems to be conflicting among various solid cancers [108]. Thomas et al. have summarized the studies on the relationship of JAK/STAT activation and prognosis [108]. In several cancers such as prostate, non-small cell lung cancers, cervical cancers, renal cell carcinoma (RCC) and glioblastoma, activation of STAT3 or STAT5 is associated with a worse prognosis; conversely, STAT3 is associated with favorable prognosis in breast cancer and in some studies in CRC and head and neck squamous cell carcinoma [108]. Different tumor biology among cancers, and the various regulatory mechanisms upstream of p-STAT3 signaling, for example, endogenous negative regulators such as the suppressor of cytokine signaling (SOCS) family, protein inhibitor of activated STAT (PIAS) proteins and the PTP family, or posttranslational modifications [109] may lead to the difference in prognosis prediction. STAT3 is highly expressed and activated in most breast cancers [110], especially in TNBC [111]. Also, high p-STAT3 levels correlated with worse outcomes in invasive breast cancers [112].
In contrast to the relatively established regulatory roles of SHP-1 in immune and hematopoietic cells [113,114], the clinical relevance of SHP-1 in cancers remain an exploratory field. There have been several studies on the expressions of SHP-1 and their significance in cancers. Some studies found decreased SHP-1 expression linked to poor outcome [115], whereas some concluded the opposite [116,117]. Immunohistochemical detection of SHP-1 has been shown to predict the outcome for localized prostate cancer; a decreased level of SHP-1 expression in prostate cancer cells is associated with a high proliferation rate and an increased risk of recurrence or clinical progression after radical prostatectomy for localized prostate cancer [115]. On the contrary, increased expression of SHP-1 is associated with local recurrence after radiotherapy for patients with nasopharyngeal carcinoma [117]. Insabato et al. found that elevated SHP-1 expressions were correlated with conventional pathologic parameters of tumor aggressiveness (such as HER2) and were associated with reduced patient survival [116]. Tao et al. found significant correlations among the infection of human papillomavirus (HPV) and the expression of SHP-1 in both condyloma acuminatum and cervical cancer, suggesting a putative role of SHP-1 in the progression of both condyloma acuminatum and cervical cancer after HPV infection [118]. Patients with an elevated SHP-1-E-cadherin axis had longer survival rate in CRC [119]. Given the relative paucity of literature on SHP-1 expressions in clinical cancer patient samples, more studies are needed to better define the prognostic roles of SHP-1 in various contexts of cancer types.
Currently, SC-compounds (SHP-1 agonists) are still in the pre-clinical stage, and none of these compounds have yet been approved as investigational new drugs (IND) for cancers. In contrast, STAT3 inhibitors that are designated based on more common strategies for inhibiting STAT3 transcriptional activity such as JAK inhibitors, STAT3 dimerization domain inhibitors, STAT3-DNA binding blockers, and antisense oligonucleotide inhibitor of STAT3 have been in various developmental stages or in clinical trials for cancer treatment (reviewed nicely in references [8,9,10,109]).

6. Conclusions and Perspectives

Loss of SHP-1 contributes to the activation of JAK/STAT3 as well as the other oncogenic pathways, and may further spark off an oncogenic feedforward loop to amplify tumorigenic signals. Hence, reactivating the function of SHP-1 phosphatase to target STAT3 can be a promising candidate for targeted cancer therapy and lay the foundation of new drug discovery. It is worth mentioning that SHP-1 has been reported as a potential target for cancer immunotherapy [120,121]. Loss of SHP-1 strongly increases the ability of Tregs to suppress inflammation [122]. In addition, blocking SHP-1 enzymatic activity by sodium stibogluconate significantly augmented the suppressor function of Treg both in vivo and ex vivo [122]. Furthermore, inhibition of SHP-1 has been shown to stabilize the conjugate formation between CD8 T-cells and antigen presenting cells [123], suggesting that SHP-1 blockage may lead to increased suppressor function. Although there is no report to elaborate the potential combination strategy of SHP-1 inhibition with other immune checkpoint inhibitors, it is still a promising and an important issue to investigate in the future.
The potential combination strategies of SHP-1 agonists and current approved chemotherapeutic drugs may also provide further insights into mechanisms of synergy and/or resistance, possibly leading to the development of clinical trials in human cancers. Analogously, abnormal SHP-1/p-STAT3 signaling can be a useful prognostic biomarker for predicting chemotherapeutic drugs’ response. These findings demonstrated an improved understanding of SHP-1/p-STAT3 interaction in promoting cancer progression and these concepts can be further translated into efficacious combination approaches to prolong the survival rate and improve the quality of life in patients. Future studies may help to delineate the impact of targeting SHP-1/STAT3 on the network among these pathways.

Author Contributions

Tzu-Ting Huang, Jung-Chen Su, Chun-Yu Liu, Chung-Wai Shiau and Kuen-Feng Chen reviewed the literature, discussed manuscript context design and wrote the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lim, C.P.; Cao, X. Structure, function, and regulation of STAT proteins. Mol. Biosyst. 2006, 2, 536–550. [Google Scholar] [CrossRef] [PubMed]
  2. Schindler, C.; Levy, D.E.; Decker, T. JAK-STAT signaling: From interferons to cytokines. J. Biol. Chem. 2007, 282, 20059–20063. [Google Scholar] [CrossRef] [PubMed]
  3. Santoni, M.; Massari, F.; Del Re, M.; Ciccarese, C.; Piva, F.; Principato, G.; Montironi, R.; Santini, D.; Danesi, R.; Tortora, G.; et al. Investigational therapies targeting signal transducer and activator of transcription 3 for the treatment of cancer. Expert Opin. Investig. Drugs 2015, 24, 809–824. [Google Scholar] [CrossRef] [PubMed]
  4. Subramaniam, A.; Shanmugam, M.K.; Perumal, E.; Li, F.; Nachiyappan, A.; Dai, X.; Swamy, S.N.; Ahn, K.S.; Kumar, A.P.; Tan, B.K.; et al. Potential role of signal transducer and activator of transcription (STAT)3 signaling pathway in inflammation, survival, proliferation and invasion of hepatocellular carcinoma. Biochim. Biophys. Acta 2013, 1835, 46–60. [Google Scholar] [CrossRef] [PubMed]
  5. Bendell, J.C.; Hong, D.S.; Burris, H.A., 3rd; Naing, A.; Jones, S.F.; Falchook, G.; Bricmont, P.; Elekes, A.; Rock, E.P.; Kurzrock, R. Phase 1, open-label, dose-escalation, and pharmacokinetic study of STAT3 inhibitor OPB-31121 in subjects with advanced solid tumors. Cancer Chemother. Pharmacol. 2014, 74, 125–130. [Google Scholar] [CrossRef] [PubMed]
  6. Hong, D.; Kurzrock, R.; Kim, Y.; Woessner, R.; Younes, A.; Nemunaitis, J.; Fowler, N.; Zhou, T.; Schmidt, J.; Jo, M.; et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci. Transl. Med. 2015, 7, 314ra185. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, X.; Yue, P.; Page, B.D.; Li, T.; Zhao, W.; Namanja, A.T.; Paladino, D.; Zhao, J.; Chen, Y.; Gunning, P.T.; et al. Orally bioavailable small-molecule inhibitor of transcription factor Stat3 regresses human breast and lung cancer xenografts. Proc. Natl. Acad. Sci. USA 2012, 109, 9623–9628. [Google Scholar] [CrossRef] [PubMed]
  8. Geiger, J.L.; Grandis, J.R.; Bauman, J.E. The STAT3 pathway as a therapeutic target in head and neck cancer: Barriers and innovations. Oral Oncol. 2016, 56, 84–92. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, X.; Crowe, P.J.; Goldstein, D.; Yang, J.L. STAT3 inhibition, a novel approach to enhancing targeted therapy in human cancers (review). Int. J. Oncol. 2012, 41, 1181–1191. [Google Scholar] [PubMed]
  10. Furtek, S.L.; Backos, D.S.; Matheson, C.J.; Reigan, P. Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem. Biol. 2016, 11, 308–318. [Google Scholar] [CrossRef] [PubMed]
  11. Siveen, K.S.; Sikka, S.; Surana, R.; Dai, X.; Zhang, J.; Kumar, A.P.; Tan, B.K.; Sethi, G.; Bishayee, A. Targeting the STAT3 signaling pathway in cancer: Role of synthetic and natural inhibitors. Biochim. Biophys. Acta 2014, 1845, 136–154. [Google Scholar] [CrossRef] [PubMed]
  12. Zhao, M.; Jiang, B.; Gao, F.H. Small molecule inhibitors of STAT3 for cancer therapy. Curr. Med. Chem. 2011, 18, 4012–4018. [Google Scholar] [CrossRef] [PubMed]
  13. Masciocchi, D.; Gelain, A.; Villa, S.; Meneghetti, F.; Barlocco, D. Signal transducer and activator of transcription 3 (STAT3): A promising target for anticancer therapy. Future Med. Chem. 2011, 3, 567–597. [Google Scholar] [CrossRef] [PubMed]
  14. Cafferkey, C.; Chau, I. Novel STAT 3 inhibitors for treating gastric cancer. Expert Opin. Investig. Drugs 2016, 25, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  15. Turkson, J.; Bowman, T.; Garcia, R.; Caldenhoven, E.; De Groot, R.P.; Jove, R. Stat3 activation by Src induces specific gene regulation and is required for cell transformation. Mol. Cell. Biol. 1998, 18, 2545–2552. [Google Scholar] [CrossRef] [PubMed]
  16. Niu, G.; Bowman, T.; Huang, M.; Shivers, S.; Reintgen, D.; Daud, A.; Chang, A.; Kraker, A.; Jove, R.; Yu, H. Roles of activated Src and Stat3 signaling in melanoma tumor cell growth. Oncogene 2002, 21, 7001–7010. [Google Scholar] [CrossRef] [PubMed]
  17. Den Hertog, J.; Ostman, A.; Bohmer, F.D. Protein tyrosine phosphatases: Regulatory mechanisms. FEBS J. 2008, 275, 831–847. [Google Scholar] [CrossRef] [PubMed]
  18. Yamada, S.; Shiono, S.; Joo, A.; Yoshimura, A. Control mechanism of JAK/STAT signal transduction pathway. FEBS Lett. 2003, 534, 190–196. [Google Scholar] [CrossRef]
  19. Tai, W.T.; Cheng, A.L.; Shiau, C.W.; Huang, H.P.; Huang, J.W.; Chen, P.J.; Chen, K.F. Signal transducer and activator of transcription 3 is a major kinase-independent target of sorafenib in hepatocellular carcinoma. J. Hepatol. 2011, 55, 1041–1048. [Google Scholar] [CrossRef] [PubMed]
  20. Beldi-Ferchiou, A.; Skouri, N.; Ben Ali, C.; Safra, I.; Abdelkefi, A.; Ladeb, S.; Mrad, K.; Ben Othman, T.; Ben Ahmed, M. Abnormal repression of SHP-1, SHP-2 and SOCS-1 transcription sustains the activation of the JAK/STAT3 pathway and the progression of the disease in multiple myeloma. PLoS ONE 2017, 12, e0174835. [Google Scholar] [CrossRef] [PubMed]
  21. Fan, L.C.; Shiau, C.W.; Tai, W.T.; Hung, M.H.; Chu, P.Y.; Hsieh, F.S.; Lin, H.; Yu, H.C.; Chen, K.F. SHP-1 is a negative regulator of epithelial-mesenchymal transition in hepatocellular carcinoma. Oncogene 2015, 34, 5252–5263. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, C.Y.; Su, J.C.; Ni, M.H.; Tseng, L.M.; Chu, P.Y.; Wang, D.S.; Tai, W.T.; Kao, Y.P.; Hung, M.H.; Shiau, C.W.; et al. Obatoclax analog SC-2001 inhibits STAT3 phosphorylation through enhancing SHP-1 expression and induces apoptosis in human breast cancer cells. Breast Cancer Res. Treat. 2014, 146, 71–84. [Google Scholar] [CrossRef] [PubMed]
  23. Su, J.C.; Mar, A.C.; Wu, S.H.; Tai, W.T.; Chu, P.Y.; Wu, C.Y.; Tseng, L.M.; Lee, T.C.; Chen, K.F.; Liu, C.Y.; et al. Disrupting VEGF-A paracrine and autocrine loops by targeting SHP-1 suppresses triple negative breast cancer metastasis. Sci. Rep. 2016, 6, 28888. [Google Scholar] [CrossRef] [PubMed]
  24. Aggarwal, B.B.; Sethi, G.; Ahn, K.S.; Sandur, S.K.; Pandey, M.K.; Kunnumakkara, A.B.; Sung, B.; Ichikawa, H. Targeting signal-transducer-and-activator-of-transcription-3 for prevention and therapy of cancer: Modern target but ancient solution. Ann. N. Y. Acad. Sci. 2006, 1091, 151–169. [Google Scholar] [CrossRef] [PubMed]
  25. Nielsen, M.; Kaestel, C.G.; Eriksen, K.W.; Woetmann, A.; Stokkedal, T.; Kaltoft, K.; Geisler, C.; Ropke, C.; Odum, N. Inhibition of constitutively activated Stat3 correlates with altered Bcl-2/Bax expression and induction of apoptosis in mycosis fungoides tumor cells. Leukemia 1999, 13, 735–738. [Google Scholar] [CrossRef] [PubMed]
  26. Cao, X.; Tay, A.; Guy, G.R.; Tan, Y.H. Activation and association of Stat3 with Src in v-Src-transformed cell lines. Mol. Cell. Biol. 1996, 16, 1595–1603. [Google Scholar] [CrossRef] [PubMed]
  27. Bromberg, J.F.; Horvath, C.M.; Besser, D.; Lathem, W.W.; Darnell, J.E., Jr. Stat3 activation is required for cellular transformation by v-src. Mol. Cell. Biol. 1998, 18, 2553–2558. [Google Scholar] [CrossRef] [PubMed]
  28. Ostman, A.; Hellberg, C.; Bohmer, F.D. Protein-tyrosine phosphatases and cancer. Nat. Rev. Cancer 2006, 6, 307–320. [Google Scholar] [CrossRef] [PubMed]
  29. David, M.; Chen, H.E.; Goelz, S.; Larner, A.C.; Neel, B.G. Differential regulation of the alpha/beta interferon-stimulated Jak/Stat pathway by the SH2 domain-containing tyrosine phosphatase SHPTP1. Mol. Cell. Biol. 1995, 15, 7050–7058. [Google Scholar] [CrossRef] [PubMed]
  30. Jiao, H.; Berrada, K.; Yang, W.; Tabrizi, M.; Platanias, L.C.; Yi, T. Direct association with and dephosphorylation of Jak2 kinase by the SH2-domain-containing protein tyrosine phosphatase SHP-1. Mol. Cell. Biol. 1996, 16, 6985–6992. [Google Scholar] [CrossRef] [PubMed]
  31. Haque, S.J.; Harbor, P.; Tabrizi, M.; Yi, T.; Williams, B.R. Protein-tyrosine phosphatase Shp-1 is a negative regulator of IL-4- and IL-13-dependent signal transduction. J. Biol. Chem. 1998, 273, 33893–33896. [Google Scholar] [CrossRef] [PubMed]
  32. Migone, T.S.; Cacalano, N.A.; Taylor, N.; Yi, T.; Waldmann, T.A.; Johnston, J.A. Recruitment of SH2-containing protein tyrosine phosphatase SHP-1 to the interleukin 2 receptor; loss of SHP-1 expression in human T-lymphotropic virus type I-transformed T cells. Proc. Natl. Acad. Sci. USA 1998, 95, 3845–3850. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, J.; Liang, X.; Niu, T.; Meng, W.; Zhao, Z.; Zhou, G.W. Crystal structure of the catalytic domain of protein-tyrosine phosphatase SHP-1. J. Biol. Chem. 1998, 273, 28199–28207. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, J.; Liu, L.; He, D.; Song, X.; Liang, X.; Zhao, Z.J.; Zhou, G.W. Crystal structure of human protein-tyrosine phosphatase SHP-1. J. Biol. Chem. 2003, 278, 6516–6520. [Google Scholar] [CrossRef] [PubMed]
  35. Thangaraju, M.; Sharma, K.; Liu, D.; Shen, S.H.; Srikant, C.B. Interdependent regulation of intracellular acidification and SHP-1 in apoptosis. Cancer Res. 1999, 59, 1649–1654. [Google Scholar] [PubMed]
  36. Lopez, F.; Esteve, J.P.; Buscail, L.; Delesque, N.; Saint-Laurent, N.; Theveniau, M.; Nahmias, C.; Vaysse, N.; Susini, C. The tyrosine phosphatase SHP-1 associates with the sst2 somatostatin receptor and is an essential component of sst2-mediated inhibitory growth signaling. J. Biol. Chem. 1997, 272, 24448–24454. [Google Scholar] [CrossRef] [PubMed]
  37. Zapata, P.D.; Ropero, R.M.; Valencia, A.M.; Buscail, L.; Lopez, J.I.; Martin-Orozco, R.M.; Prieto, J.C.; Angulo, J.; Susini, C.; Lopez-Ruiz, P.; et al. Autocrine regulation of human prostate carcinoma cell proliferation by somatostatin through the modulation of the SH2 domain containing protein tyrosine phosphatase (SHP)-1. J. Clin. Endocrinol. Metab. 2002, 87, 915–926. [Google Scholar] [CrossRef] [PubMed]
  38. Delibrias, C.C.; Floettmann, J.E.; Rowe, M.; Fearon, D.T. Downregulated expression of SHP-1 in Burkitt lymphomas and germinal center B lymphocytes. J. Exp. Med. 1997, 186, 1575–1583. [Google Scholar] [CrossRef] [PubMed]
  39. Wu, C.; Sun, M.; Liu, L.; Zhou, G.W. The function of the protein tyrosine phosphatase SHP-1 in cancer. Gene 2003, 306, 1–12. [Google Scholar] [CrossRef]
  40. Chim, C.S.; Fung, T.K.; Cheung, W.C.; Liang, R.; Kwong, Y.L. SOCS1 and SHP1 hypermethylation in multiple myeloma: Implications for epigenetic activation of the Jak/STAT pathway. Blood 2004, 103, 4630–4635. [Google Scholar] [CrossRef] [PubMed]
  41. Zhang, Q.; Wang, H.Y.; Marzec, M.; Raghunath, P.N.; Nagasawa, T.; Wasik, M.A. STAT3- and DNA methyltransferase 1-mediated epigenetic silencing of SHP-1 tyrosine phosphatase tumor suppressor gene in malignant T lymphocytes. Proc. Natl. Acad. Sci. USA 2005, 102, 6948–6953. [Google Scholar] [CrossRef] [PubMed]
  42. Shaulian, E.; Karin, M. AP-1 as a regulator of cell life and death. Nat. Cell Biol. 2002, 4, E131–E136. [Google Scholar] [CrossRef] [PubMed]
  43. Zugowski, C.; Lieder, F.; Muller, A.; Gasch, J.; Corvinus, F.M.; Moriggl, R.; Friedrich, K. STAT3 controls matrix metalloproteinase-1 expression in colon carcinoma cells by both direct and AP-1-mediated interaction with the MMP-1 promoter. Biol. Chem. 2011, 392, 449–459. [Google Scholar] [CrossRef] [PubMed]
  44. Du, Q.; Geller, D.A. Cross-Regulation Between Wnt and NF-kappaB Signaling Pathways. Forum Immunopathol. Dis. Ther. 2010, 1, 155–181. [Google Scholar] [CrossRef]
  45. Kawada, M.; Seno, H.; Uenoyama, Y.; Sawabu, T.; Kanda, N.; Fukui, H.; Shimahara, Y.; Chiba, T. Signal transducers and activators of transcription 3 activation is involved in nuclear accumulation of beta-catenin in colorectal cancer. Cancer Res. 2006, 66, 2913–2917. [Google Scholar] [CrossRef] [PubMed]
  46. Yan, S.; Zhou, C.; Zhang, W.; Zhang, G.; Zhao, X.; Yang, S.; Wang, Y.; Lu, N.; Zhu, H.; Xu, N. Beta-Catenin/TCF pathway upregulates STAT3 expression in human esophageal squamous cell carcinoma. Cancer Lett. 2008, 271, 85–97. [Google Scholar] [CrossRef] [PubMed]
  47. Fragoso, M.A.; Patel, A.K.; Nakamura, R.E.; Yi, H.; Surapaneni, K.; Hackam, A.S. The Wnt/beta-catenin pathway cross-talks with STAT3 signaling to regulate survival of retinal pigment epithelium cells. PLoS ONE 2012, 7, e46892. [Google Scholar] [CrossRef] [PubMed]
  48. Hao, J.; Li, T.G.; Qi, X.; Zhao, D.F.; Zhao, G.Q. WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev. Biol. 2006, 290, 81–91. [Google Scholar] [CrossRef] [PubMed]
  49. Ye, S.; Zhang, D.; Cheng, F.; Wilson, D.; Mackay, J.; He, K.; Ban, Q.; Lv, F.; Huang, S.; Liu, D.; et al. Wnt/beta-catenin and LIF-Stat3 signaling pathways converge on Sp5 to promote mouse embryonic stem cell self-renewal. J. Cell Sci. 2016, 129, 269–276. [Google Scholar] [CrossRef] [PubMed]
  50. Fan, Y.; Mao, R.; Yang, J. NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 2013, 4, 176–185. [Google Scholar] [CrossRef] [PubMed]
  51. Grivennikov, S.I.; Karin, M. Dangerous liaisons: STAT3 and NF-κB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010, 21, 11–19. [Google Scholar] [CrossRef] [PubMed]
  52. Bollrath, J.; Greten, F.R. IKK/NF-kappaB and STAT3 pathways: Central signalling hubs in inflammation-mediated tumour promotion and metastasis. EMBO Rep. 2009, 10, 1314–1319. [Google Scholar] [CrossRef] [PubMed]
  53. Forget, G.; Siminovitch, K.A.; Brochu, S.; Rivest, S.; Radzioch, D.; Olivier, M. Role of host phosphotyrosine phosphatase SHP-1 in the development of murine leishmaniasis. Eur. J. Immunol. 2001, 31, 3185–3196. [Google Scholar] [CrossRef]
  54. Forget, G.; Gregory, D.J.; Whitcombe, L.A.; Olivier, M. Role of host protein tyrosine phosphatase SHP-1 in Leishmania donovani-induced inhibition of nitric oxide production. Infect. Immun. 2006, 74, 6272–6279. [Google Scholar] [CrossRef] [PubMed]
  55. Massa, P.T.; Wu, C. Increased inducible activation of NF-κB and responsive genes in astrocytes deficient in the protein tyrosine phosphatase SHP-1. J. Interferon Cytokine Res. 1998, 18, 499–507. [Google Scholar] [CrossRef] [PubMed]
  56. Duchesne, C.; Charland, S.; Asselin, C.; Nahmias, C.; Rivard, N. Negative regulation of beta-catenin signaling by tyrosine phosphatase SHP-1 in intestinal epithelial cells. J. Biol. Chem. 2003, 278, 14274–14283. [Google Scholar] [CrossRef] [PubMed]
  57. Simpson, D.; Keating, G.M. Sorafenib: In hepatocellular carcinoma. Drugs 2008, 68, 251–258. [Google Scholar] [CrossRef] [PubMed]
  58. Yang, F.; Van Meter, T.E.; Buettner, R.; Hedvat, M.; Liang, W.; Kowolik, C.M.; Mepani, N.; Mirosevich, J.; Nam, S.; Chen, M.Y.; 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. [Google Scholar] [CrossRef] [PubMed]
  59. Huang, S.; Sinicrope, F.A. Sorafenib inhibits STAT3 activation to enhance TRAIL-mediated apoptosis in human pancreatic cancer cells. Mol. Cancer Ther. 2010, 9, 742–750. [Google Scholar] [CrossRef] [PubMed]
  60. Yang, F.; Brown, C.; Buettner, R.; Hedvat, M.; Starr, R.; Scuto, A.; Schroeder, A.; Jensen, M.; Jove, R. Sorafenib induces growth arrest and apoptosis of human glioblastoma cells through the dephosphorylation of signal transducers and activators of transcription 3. Mol. Cancer Ther. 2010, 9, 953–962. [Google Scholar] [CrossRef] [PubMed]
  61. Chai, H.; Luo, A.Z.; Weerasinghe, P.; Brown, R.E. Sorafenib downregulates ERK/Akt and STAT3 survival pathways and induces apoptosis in a human neuroblastoma cell line. Int. J. Clin. Exp. Pathol. 2010, 3, 408–415. [Google Scholar] [PubMed]
  62. Zhao, W.; Zhang, T.; Qu, B.; Wu, X.; Zhu, X.; Meng, F.; Gu, Y.; Shu, Y.; Shen, Y.; Sun, Y.; et al. Sorafenib induces apoptosis in HL60 cells by inhibiting Src kinase-mediated STAT3 phosphorylation. Anti Cancer Drugs 2011, 22, 79–88. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, K.F.; Tai, W.T.; Liu, T.H.; Huang, H.P.; Lin, Y.C.; Shiau, C.W.; Li, P.K.; Chen, P.J.; Cheng, A.L. Sorafenib overcomes TRAIL resistance of hepatocellular carcinoma cells through the inhibition of STAT3. Clin. Cancer Res. 2010, 16, 5189–5199. [Google Scholar] [CrossRef] [PubMed]
  64. Chao, T.I.; Tai, W.T.; Hung, M.H.; Tsai, M.H.; Chen, M.H.; Chang, M.J.; Shiau, C.W.; Chen, K.F. A combination of sorafenib and SC-43 is a synergistic SHP-1 agonist duo to advance hepatocellular carcinoma therapy. Cancer Lett. 2016, 371, 205–213. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, K.F.; Tai, W.T.; Hsu, C.Y.; Huang, J.W.; Liu, C.Y.; Chen, P.J.; Kim, I.; Shiau, C.W. Blockade of STAT3 activation by sorafenib derivatives through enhancing SHP-1 phosphatase activity. Eur. J. Med. Chem. 2012, 55, 220–227. [Google Scholar] [CrossRef] [PubMed]
  66. Adnane, L.; Trail, P.A.; Taylor, I.; Wilhelm, S.M. Sorafenib (BAY 43-9006, Nexavar), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 2006, 407, 597–612. [Google Scholar] [PubMed]
  67. Liu, L.; Cao, Y.; Chen, C.; Zhang, X.; McNabola, A.; Wilkie, D.; Wilhelm, S.; Lynch, M.; Carter, C. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006, 66, 11851–11858. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, C.Y.; Tseng, L.M.; Su, J.C.; Chang, K.C.; Chu, P.Y.; Tai, W.T.; Shiau, C.W.; Chen, K.F. Novel sorafenib analogues induce apoptosis through SHP-1 dependent STAT3 inactivation in human breast cancer cells. Breast Cancer Res. BCR 2013, 15, R63. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, C.Y.; Su, J.C.; Huang, T.T.; Chu, P.Y.; Huang, C.T.; Wang, W.L.; Lee, C.H.; Lau, K.Y.; Tsai, W.C.; Yang, H.P.; et al. Sorafenib analogue SC-60 induces apoptosis through the SHP-1/STAT3 pathway and enhances docetaxel cytotoxicity in triple-negative breast cancer cells. Mol. Oncol. 2017, 11, 266–279. [Google Scholar] [CrossRef] [PubMed]
  70. Tai, W.T.; Shiau, C.W.; Chen, P.J.; Chu, P.Y.; Huang, H.P.; Liu, C.Y.; Huang, J.W.; Chen, K.F. Discovery of novel Src homology region 2 domain-containing phosphatase 1 agonists from sorafenib for the treatment of hepatocellular carcinoma. Hepatology 2014, 59, 190–201. [Google Scholar] [CrossRef] [PubMed]
  71. Hu, M.H.; Chen, L.J.; Chen, Y.L.; Tsai, M.S.; Shiau, C.W.; Chao, T.I.; Liu, C.Y.; Kao, J.H.; Chen, K.F. Targeting SHP-1-STAT3 signaling: A promising therapeutic approach for the treatment of cholangiocarcinoma. Oncotarget 2017. [Google Scholar] [CrossRef] [PubMed]
  72. Tai, W.T.; Shiau, C.W.; Li, Y.S.; Chen, Y.L.; Chu, P.Y.; Huang, J.W.; Hsu, C.Y.; Hsu, Y.C.; Chen, P.J.; Chen, K.F. SC-60, a dimer-based sorafenib derivative, shows a better anti-hepatocellular carcinoma effect than sorafenib in a preclinical hepatocellular carcinoma model. Mol. Cancer Ther. 2014, 13, 27–36. [Google Scholar] [CrossRef] [PubMed]
  73. Fan, L.C.; Teng, H.W.; Shiau, C.W.; Tai, W.T.; Hung, M.H.; Yang, S.H.; Jiang, J.K.; Chen, K.F. Pharmacological Targeting SHP-1-STAT3 Signaling Is a Promising Therapeutic Approach for the Treatment of Colorectal Cancer. Neoplasia 2015, 17, 687–696. [Google Scholar] [CrossRef] [PubMed]
  74. Fan, L.C.; Teng, H.W.; Shiau, C.W.; Lin, H.; Hung, M.H.; Chen, Y.L.; Huang, J.W.; Tai, W.T.; Yu, H.C.; Chen, K.F. SHP-1 is a target of regorafenib in colorectal cancer. Oncotarget 2014, 5, 6243–6251. [Google Scholar] [CrossRef] [PubMed]
  75. Tai, W.T.; Chu, P.Y.; Shiau, C.W.; Chen, Y.L.; Li, Y.S.; Hung, M.H.; Chen, L.J.; Chen, P.L.; Su, J.C.; Lin, P.Y.; et al. STAT3 mediates regorafenib-induced apoptosis in hepatocellular carcinoma. Clin. Cancer Res. 2014, 20, 5768–5776. [Google Scholar] [CrossRef] [PubMed]
  76. Tai, W.T.; Cheng, A.L.; Shiau, C.W.; Liu, C.Y.; Ko, C.H.; Lin, M.W.; Chen, P.J.; Chen, K.F. Dovitinib induces apoptosis and overcomes sorafenib resistance in hepatocellular carcinoma through SHP-1-mediated inhibition of STAT3. Mol. Cancer Ther. 2012, 11, 452–463. [Google Scholar] [CrossRef] [PubMed]
  77. Tai, W.T.; Shiau, C.W.; Li, Y.S.; Chang, C.W.; Huang, J.W.; Hsueh, T.T.; Yu, H.C.; Chen, K.F. Nintedanib (BIBF-1120) inhibits hepatocellular carcinoma growth independent of angiokinase activity. J. Hepatol. 2014, 61, 89–97. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, J.H.; Chiang, S.Y.; Nam, D.; Chung, W.S.; Lee, J.; Na, Y.S.; Sethi, G.; Ahn, K.S. Capillarisin inhibits constitutive and inducible STAT3 activation through induction of SHP-1 and SHP-2 tyrosine phosphatases. Cancer Lett. 2014, 345, 140–148. [Google Scholar] [CrossRef] [PubMed]
  79. Kim, S.H.; Ahn, K.S.; Jeong, S.J.; Kwon, T.R.; Jung, J.H.; Yun, S.M.; Han, I.; Lee, S.G.; Kim, D.K.; Kang, M.; et al. Janus activated kinase 2/signal transducer and activator of transcription 3 pathway mediates icariside II-induced apoptosis in U266 multiple myeloma cells. Eur. J. Pharmacol. 2011, 654, 10–16. [Google Scholar] [CrossRef] [PubMed]
  80. Pandey, M.K.; Sung, B.; Aggarwal, B.B. Betulinic acid suppresses STAT3 activation pathway through induction of protein tyrosine phosphatase SHP-1 in human multiple myeloma cells. Int. J. Cancer 2010, 127, 282–292. [Google Scholar] [CrossRef] [PubMed]
  81. Rhee, Y.H.; Jeong, S.J.; Lee, H.J.; Lee, H.J.; Koh, W.; Jung, J.H.; Kim, S.H.; Sung-Hoon, K. Inhibition of STAT3 signaling and induction of SHP1 mediate antiangiogenic and antitumor activities of ergosterol peroxide in U266 multiple myeloma cells. BMC Cancer 2012, 12, 28. [Google Scholar] [CrossRef] [PubMed]
  82. Jung, J.H.; Yun, M.; Choo, E.J.; Kim, S.H.; Jeong, M.S.; Jung, D.B.; Lee, H.; Kim, E.O.; Kato, N.; Kim, B.; et al. A derivative of epigallocatechin-3-gallate induces apoptosis via SHP-1-mediated suppression of BCR-ABL and STAT3 signalling in chronic myelogenous leukaemia. Br. J. Pharmacol. 2015, 172, 3565–3578. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, J.C.; Ahn, K.S.; Jeong, S.J.; Jung, J.H.; Kwon, T.R.; Rhee, Y.H.; Kim, S.H.; Kim, S.Y.; Yoon, H.J.; Zhu, S.; et al. Signal transducer and activator of transcription 3 pathway mediates genipin-induced apoptosis in U266 multiple myeloma cells. J. Cell. Biochem. 2011, 112, 1552–1562. [Google Scholar] [CrossRef] [PubMed]
  84. Kunnumakkara, A.B.; Nair, A.S.; Sung, B.; Pandey, M.K.; Aggarwal, B.B. Boswellic acid blocks signal transducers and activators of transcription 3 signaling, proliferation, and survival of multiple myeloma via the protein tyrosine phosphatase SHP-1. Mol. Cancer Res. MCR 2009, 7, 118–128. [Google Scholar] [CrossRef] [PubMed]
  85. Baek, S.H.; Lee, J.H.; Kim, C.; Ko, J.H.; Ryu, S.H.; Lee, S.G.; Yang, W.M.; Um, J.Y.; Chinnathambi, A.; Alharbi, S.A.; et al. Ginkgolic Acid C 17:1, Derived from Ginkgo biloba Leaves, Suppresses Constitutive and Inducible STAT3 Activation through Induction of PTEN and SHP-1 Tyrosine Phosphatase. Molecules 2017. [Google Scholar] [CrossRef] [PubMed]
  86. Subramaniam, A.; Shanmugam, M.K.; Ong, T.H.; Li, F.; Perumal, E.; Chen, L.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; et al. Emodin inhibits growth and induces apoptosis in an orthotopic hepatocellular carcinoma model by blocking activation of STAT3. Br. J. Pharmacol. 2013, 170, 807–821. [Google Scholar] [CrossRef] [PubMed]
  87. Rajendran, P.; Li, F.; Manu, K.A.; Shanmugam, M.K.; Loo, S.Y.; Kumar, A.P.; Sethi, G. gamma-Tocotrienol is a novel inhibitor of constitutive and inducible STAT3 signalling pathway in human hepatocellular carcinoma: Potential role as an antiproliferative, pro-apoptotic and chemosensitizing agent. Br. J. Pharmacol. 2011, 163, 283–298. [Google Scholar] [CrossRef] [PubMed]
  88. Rajendran, P.; Li, F.; Shanmugam, M.K.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J. Cell. Physiol. 2012, 227, 2184–2195. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, T.; Li, S.; Li, J.; Yin, F.; Hua, Y.; Wang, Z.; Lin, B.; Wang, H.; Zou, D.; Zhou, Z.; et al. Natural product pectolinarigenin inhibits osteosarcoma growth and metastasis via SHP-1-mediated STAT3 signaling inhibition. Cell Death Dis. 2016, 7, e2421. [Google Scholar] [CrossRef] [PubMed]
  90. Shanmugam, M.K.; Rajendran, P.; Li, F.; Kim, C.; Sikka, S.; Siveen, K.S.; Kumar, A.P.; Ahn, K.S.; Sethi, G. Abrogation of STAT3 signaling cascade by zerumbone inhibits proliferation and induces apoptosis in renal cell carcinoma xenograft mouse model. Mol. Carcinogenes. 2015, 54, 971–985. [Google Scholar] [CrossRef] [PubMed]
  91. Song, S.; Su, Z.; Xu, H.; Niu, M.; Chen, X.; Min, H.; Zhang, B.; Sun, G.; Xie, S.; Wang, H.; et al. Luteolin selectively kills STAT3 highly activated gastric cancer cells through enhancing the binding of STAT3 to SHP-1. Cell Death Dis. 2017, 8, e2612. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, K.F.; Chen, H.L.; Shiau, C.W.; Liu, C.Y.; Chu, P.Y.; Tai, W.T.; Ichikawa, K.; Chen, P.J.; Cheng, A.L. Sorafenib and its derivative SC-49 sensitize hepatocellular carcinoma cells to CS-1008, a humanized anti-TNFRSF10B (DR5) antibody. Br. J. Pharmacol. 2013, 168, 658–672. [Google Scholar] [CrossRef] [PubMed]
  93. Huang, C.Y.; Tai, W.T.; Hsieh, C.Y.; Hsu, W.M.; Lai, Y.J.; Chen, L.J.; Shiau, C.W.; Chen, K.F. A sorafenib derivative and novel SHP-1 agonist, SC-59, acts synergistically with radiotherapy in hepatocellular carcinoma cells through inhibition of STAT3. Cancer Lett. 2014, 349, 136–143. [Google Scholar] [CrossRef] [PubMed]
  94. Su, J.C.; Chiang, H.C.; Tseng, P.H.; Tai, W.T.; Hsu, C.Y.; Li, Y.S.; Huang, J.W.; Ko, C.H.; Lin, M.W.; Chu, P.Y.; et al. RFX-1-dependent activation of SHP-1 inhibits STAT3 signaling in hepatocellular carcinoma cells. Carcinogenesis 2014, 35, 2807–2814. [Google Scholar] [CrossRef] [PubMed]
  95. Su, J.C.; Tseng, P.H.; Wu, S.H.; Hsu, C.Y.; Tai, W.T.; Li, Y.S.; Chen, I.T.; Liu, C.Y.; Chen, K.F.; Shiau, C.W. SC-2001 overcomes STAT3-mediated sorafenib resistance through RFX-1/SHP-1 activation in hepatocellular carcinoma. Neoplasia 2014, 16, 595–605. [Google Scholar] [CrossRef] [PubMed]
  96. Olweny, C.L.; Toya, T.; Katongole-Mbidde, E.; Mugerwa, J.; Kyalwazi, S.K.; Cohen, H. Treatment of hepatocellular carcinoma with adriamycin. Preliminary communication. Cancer 1975, 36, 1250–1257. [Google Scholar] [CrossRef]
  97. Stalmeier, P.F.; van Tol-Geerdink, J.J.; van Lin, E.N.; Schimmel, E.; Huizenga, H.; van Daal, W.A.; Leer, J.W. Doctors’ and patients’ preferences for participation and treatment in curative prostate cancer radiotherapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2007, 25, 3096–3100. [Google Scholar] [CrossRef] [PubMed]
  98. Yeo, W.; Mok, T.S.; Zee, B.; Leung, T.W.; Lai, P.B.; Lau, W.Y.; Koh, J.; Mo, F.K.; Yu, S.C.; Chan, A.T.; et al. A randomized phase III study of doxorubicin versus cisplatin/interferon α-2b/doxorubicin/fluorouracil (PIAF) combination chemotherapy for unresectable hepatocellular carcinoma. J. Natl. Cancer Inst. 2005, 97, 1532–1538. [Google Scholar] [CrossRef] [PubMed]
  99. Choi, T.K.; Lee, N.W.; Wong, J. Chemotherapy for advanced hepatocellular carcinoma. Adriamycin versus quadruple chemotherapy. Cancer 1984, 53, 401–405. [Google Scholar] [CrossRef]
  100. Melia, W.M.; Johnson, P.J.; Williams, R. Induction of remission in hepatocellular carcinoma. A comparison of VP 16 with adriamycin. Cancer 1983, 51, 206–210. [Google Scholar] [CrossRef]
  101. Falkson, G.; Moertel, C.G.; Lavin, P.; Pretorius, F.J.; Carbone, P.P. Chemotherapy studies in primary liver cancer: A prospective randomized clinical trial. Cancer 1978, 42, 2149–2156. [Google Scholar] [CrossRef]
  102. Bachawal, S.V.; Wali, V.B.; Sylvester, P.W. Combined gamma-tocotrienol and erlotinib/gefitinib treatment suppresses Stat and Akt signaling in murine mammary tumor cells. Anticancer Res. 2010, 30, 429–437. [Google Scholar] [PubMed]
  103. Huang, C.Y.; Tai, W.T.; Wu, S.Y.; Shih, C.T.; Chen, M.H.; Tsai, M.H.; Kuo, C.W.; Shiau, C.W.; Hung, M.H.; Chen, K.F. Dovitinib Acts As a Novel Radiosensitizer in Hepatocellular Carcinoma by Targeting SHP-1/STAT3 Signaling. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 761–771. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, K.F.; Chen, H.L.; Liu, C.Y.; Tai, W.T.; Ichikawa, K.; Chen, P.J.; Cheng, A.L. Dovitinib sensitizes hepatocellular carcinoma cells to TRAIL and tigatuzumab, a novel anti-DR5 antibody, through SHP-1-dependent inhibition of STAT3. Biochem. Pharmacol. 2012, 83, 769–777. [Google Scholar] [CrossRef] [PubMed]
  105. Al-Jamal, H.A.; Mat Jusoh, S.A.; Hassan, R.; Johan, M.F. Enhancing SHP-1 expression with 5-azacytidine may inhibit STAT3 activation and confer sensitivity in lestaurtinib (CEP-701)-resistant FLT3-ITD positive acute myeloid leukemia. BMC Cancer 2015, 15, 869. [Google Scholar] [CrossRef] [PubMed]
  106. Kong, J.; Kong, F.; Gao, J.; Zhang, Q.; Dong, S.; Gu, F.; Ke, S.; Pan, B.; Shen, Q.; Sun, H.; et al. YC-1 enhances the anti-tumor activity of sorafenib through inhibition of signal transducer and activator of transcription 3 (STAT3) in hepatocellular carcinoma. Mol. Cancer 2014, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  107. Sandur, S.K.; Pandey, M.K.; Sung, B.; Aggarwal, B.B. 5-hydroxy-2-methyl-1,4-naphthoquinone, a vitamin K3 analogue, suppresses STAT3 activation pathway through induction of protein tyrosine phosphatase, SHP-1: Potential role in chemosensitization. Mol. Cancer Res. MCR 2010, 8, 107–118. [Google Scholar] [CrossRef] [PubMed]
  108. Thomas, S.J.; Snowden, J.A.; Zeidler, M.P.; Danson, S.J. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br. J. Cancer 2015, 113, 365–371. [Google Scholar] [CrossRef] [PubMed]
  109. Chai, E.Z.; Shanmugam, M.K.; Arfuso, F.; Dharmarajan, A.; Wang, C.; Kumar, A.P.; Samy, R.P.; Lim, L.H.; Wang, L.; Goh, B.C.; et al. Targeting transcription factor STAT3 for cancer prevention and therapy. Pharmacol. Ther. 2016, 162, 86–97. [Google Scholar] [CrossRef] [PubMed]
  110. Kim, S.R.; Seo, H.S.; Choi, H.S.; Cho, S.G.; Kim, Y.K.; Hong, E.H.; Shin, Y.C.; Ko, S.G. Trichosanthes kirilowii Ethanol Extract and Cucurbitacin D Inhibit Cell Growth and Induce Apoptosis through Inhibition of STAT3 Activity in Breast Cancer Cells. Evid. Based Complement. Altern. Med. 2013, 2013, 975350. [Google Scholar] [CrossRef] [PubMed]
  111. Walker, S.R.; Xiang, M.; Frank, D.A. Distinct roles of STAT3 and STAT5 in the pathogenesis and targeted therapy of breast cancer. Mol. Cell. Endocrinol. 2014, 382, 616–621. [Google Scholar] [CrossRef] [PubMed]
  112. Garcia, R.; Bowman, T.L.; Niu, G.; Yu, H.; Minton, S.; Muro-Cacho, C.A.; Cox, C.E.; Falcone, R.; Fairclough, R.; Parsons, S.; et al. Constitutive activation of Stat3 by the Src and JAK tyrosine kinases participates in growth regulation of human breast carcinoma cells. Oncogene 2001, 20, 2499–2513. [Google Scholar] [CrossRef] [PubMed]
  113. Sharma, Y.; Bashir, S.; Bhardwaj, P.; Ahmad, A.; Khan, F. Protein tyrosine phosphatase SHP-1: Resurgence as new drug target for human autoimmune disorders. Immunol. Res. 2016, 64, 804–819. [Google Scholar] [CrossRef] [PubMed]
  114. Paling, N.R.; Welham, M.J. Tyrosine phosphatase SHP-1 acts at different stages of development to regulate hematopoiesis. Blood 2005, 105, 4290–4297. [Google Scholar] [CrossRef] [PubMed]
  115. Tassidis, H.; Brokken, L.J.; Jirstrom, K.; Ehrnstrom, R.; Ponten, F.; Ulmert, D.; Bjartell, A.; Harkonen, P.; Wingren, A.G. Immunohistochemical detection of tyrosine phosphatase SHP-1 predicts outcome after radical prostatectomy for localized prostate cancer. Int. J. Cancer 2010, 126, 2296–2307. [Google Scholar] [CrossRef] [PubMed]
  116. Insabato, L.; Amelio, I.; Quarto, M.; Zannetti, A.; Tolino, F.; de Mauro, G.; Cerchia, L.; Riccio, P.; Baumhoer, D.; Condorelli, G.; et al. Elevated expression of the tyrosine phosphatase SHP-1 defines a subset of high-grade breast tumors. Oncology 2009, 77, 378–384. [Google Scholar] [CrossRef] [PubMed]
  117. Peng, G.; Cao, R.; Xue, J.; Li, P.; Zou, Z.; Huang, J.; Ding, Q. Increased expression of SHP-1 is associated with local recurrence after radiotherapy in patients with nasopharyngeal carcinoma. Radiol. Oncol. 2014, 48, 40–49. [Google Scholar] [CrossRef] [PubMed]
  118. Tao, X.H.; Shen, J.G.; Pan, W.L.; Dong, Y.E.; Meng, Q.; Honn, K.V.; Jin, R. Significance of SHP-1 and SHP-2 expression in human papillomavirus infected Condyloma acuminatum and cervical cancer. Pathol. Oncol. Res. POR 2008, 14, 365–371. [Google Scholar] [CrossRef] [PubMed]
  119. Fan, L.C.; Teng, H.W.; Shiau, C.W.; Tai, W.T.; Hung, M.H.; Yang, S.H.; Jiang, J.K.; Chen, K.F. Regorafenib (Stivarga) pharmacologically targets epithelial-mesenchymal transition in colorectal cancer. Oncotarget 2016, 7, 64136–64147. [Google Scholar] [CrossRef] [PubMed]
  120. Aschauer, L.; Muller, P.A. Novel targets and interaction partners of mutant p53 Gain-Of-Function. Biochem. Soc. Trans. 2016, 44, 460–466. [Google Scholar] [CrossRef] [PubMed]
  121. Watson, H.A.; Wehenkel, S.; Matthews, J.; Ager, A. SHP-1: The next checkpoint target for cancer immunotherapy? Biochem. Soc. Trans. 2016, 44, 356–362. [Google Scholar] [CrossRef] [PubMed]
  122. Iype, T.; Sankarshanan, M.; Mauldin, I.S.; Mullins, D.W.; Lorenz, U. The protein tyrosine phosphatase SHP-1 modulates the suppressive activity of regulatory T cells. J. Immunol. 2010, 185, 6115–6127. [Google Scholar] [CrossRef] [PubMed]
  123. Sathish, J.G.; Dolton, G.; Leroy, F.G.; Matthews, R.J. Loss of Src homology region 2 domain-containing protein tyrosine phosphatase-1 increases CD8+ T cell-APC conjugate formation and is associated with enhanced in vivo CTL function. J. Immunol. 2007, 178, 330–337. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SH2 domain-containing protein tyrosine phosphatase 1 (SHP-1) inhibits signal transducer and activator of transcription 3 (STAT3) signaling pathways. In cancer cells, activated JAKs phosphorylates STAT3 (1), resulting in the translocation of activated STAT3 (p-STAT3) dimers to the nucleus as well as the activation of STAT3-regulated cellular proliferation and survival (cyclin D1, survivin, c-Myc and Mcl1), metastasis (MMPs, Rho and Rac) and angiogenesis (VEGFA) (2). Activated STAT3 also forms complexes with DNA methyltransferase 1 (DNMT1) at the promoter region of SHP-1 gene (3) to silence its transcription (4), leading to a decrement in protein levels (5). Enhancing SHP-1 activity by SHP-1 agonists (6) can directly dephosphorylate STAT3 (7) or its upstream JAKs (8) to decrease the p-STAT3 proteins (9) accompanied with the blockage of the STAT3-mediated cellular signaling pathways. Abbreviations: DNA methyltransferase 1 (DNMT1); Janus associated kinase (JAK); Matrix metalloproteinases (MMPs); Receptor tyrosine kinase (RTK); SHP-1 (SH2 domain-containing protein tyrosine phosphatase 1); Signal transducer and activator of transcription 3 (STAT3); Vascular endothelial growth factor (VEGF).
Figure 1. SH2 domain-containing protein tyrosine phosphatase 1 (SHP-1) inhibits signal transducer and activator of transcription 3 (STAT3) signaling pathways. In cancer cells, activated JAKs phosphorylates STAT3 (1), resulting in the translocation of activated STAT3 (p-STAT3) dimers to the nucleus as well as the activation of STAT3-regulated cellular proliferation and survival (cyclin D1, survivin, c-Myc and Mcl1), metastasis (MMPs, Rho and Rac) and angiogenesis (VEGFA) (2). Activated STAT3 also forms complexes with DNA methyltransferase 1 (DNMT1) at the promoter region of SHP-1 gene (3) to silence its transcription (4), leading to a decrement in protein levels (5). Enhancing SHP-1 activity by SHP-1 agonists (6) can directly dephosphorylate STAT3 (7) or its upstream JAKs (8) to decrease the p-STAT3 proteins (9) accompanied with the blockage of the STAT3-mediated cellular signaling pathways. Abbreviations: DNA methyltransferase 1 (DNMT1); Janus associated kinase (JAK); Matrix metalloproteinases (MMPs); Receptor tyrosine kinase (RTK); SHP-1 (SH2 domain-containing protein tyrosine phosphatase 1); Signal transducer and activator of transcription 3 (STAT3); Vascular endothelial growth factor (VEGF).
Ijms 18 01234 g001

Share and Cite

MDPI and ACS Style

Huang, T.-T.; Su, J.-C.; Liu, C.-Y.; Shiau, C.-W.; Chen, K.-F. Alteration of SHP-1/p-STAT3 Signaling: A Potential Target for Anticancer Therapy. Int. J. Mol. Sci. 2017, 18, 1234. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061234

AMA Style

Huang T-T, Su J-C, Liu C-Y, Shiau C-W, Chen K-F. Alteration of SHP-1/p-STAT3 Signaling: A Potential Target for Anticancer Therapy. International Journal of Molecular Sciences. 2017; 18(6):1234. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061234

Chicago/Turabian Style

Huang, Tzu-Ting, Jung-Chen Su, Chun-Yu Liu, Chung-Wai Shiau, and Kuen-Feng Chen. 2017. "Alteration of SHP-1/p-STAT3 Signaling: A Potential Target for Anticancer Therapy" International Journal of Molecular Sciences 18, no. 6: 1234. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061234

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop