We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Signal transducer and activator of transcription 3 (STAT3): a promising target for anticancer therapy

    Daniela Masciocchi

    Dipartimento di Scienze Farmaceutiche ‘P. Pratesi’, Università degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy

    , ,
    Stefania Villa

    Dipartimento di Scienze Farmaceutiche ‘P. Pratesi’, Università degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy

    ,
    Fiorella Meneghetti

    Dipartimento di Scienze Farmaceutiche ‘P. Pratesi’, Università degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy

    &
    Daniela Barlocco

    Dipartimento di Scienze Farmaceutiche ‘P. Pratesi’, Università degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy

    Published Online:https://doi.org/10.4155/fmc.11.22

    Signal transducer and activator of transcription 3 (STAT3) is an oncogenic protein whose inhibition is sought for the prevention and treatment of cancer. In this review, the validated therapeutic strategy to block aberrant activity of STAT3 in many tumor cell lines is evaluated by presenting the most promising inhibitors to date. The compounds are discussed in classes based on their different mechanisms of action, which are critically explained. In addition, their future clinical development as anticancer agents is considered. Furthermore, the efforts devoted to the comprehension of the structure–activity relationships and to the identification of the biological effects are brought to attention. The synthetic and technological approaches recently developed to overcome the difficulties in the obtainment of clinically suitable drugs are also presented.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Darnell JE Jr. STATs and gene regulation. Science277,1630–1635 (1997).
    • Bromberg J, Darnell JE. The role of STATs in transcriptional control and their impact on cellular function. Oncogene19,2468–2473 (2000).
    • Darnell JE Jr. Transcription factors as targets for cancer therapy. Nat. Rev. Cancer2,740–749 (2002).▪ Underlines the role of signal transducer and activator of transcription (STAT) as a target for cancer therapy.
    • Yu H, Jove R. The STATs of cancer – new molecular targets come of age. Nat. Rev. Cancer4,97–105 (2004).
    • Krebs DL, Hilton DJ. SOCS: physiological suppressors of cytokine signaling. J. Cell Sci.113(Pt. 16),2813–2819 (2000).
    • Isomoto H, Mott JL, Kobayashi S. Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing. Gastroenterology132(1),384–396 (2007).
    • Shuai K. Modulation of STAT signaling by STAT-interacting proteins. Oncogene21,2638–2644 (2000).
    • Shuai K, Liu B. Regulation of gene-activation pathways by PIAS proteins in the immune system. Nat. Rev. Immunol.5,593–605 (2005).
    • Yasukawa H, Sasaki A, Yoshimura A. Negative regulation of cytokine signaling pathways. Annu. Rev. Immunol.18,143–164 (2000).
    • 10  Daino H, Matsumura I, Takada K et al. Induction of apoptosis by extracellular ubiquitin in human hematopoietic cells: possible involvement of STAT3 degradation by proteasome pathway in interleukin 6-dependent hematopoietic cells. Blood95,2577–2585 (2000).
    • 11  Yamada S, Shiono S, Joo A, Yoshimura A. Control mechanism of JAK/STAT signal transduction pathway. FEBS Lett.534,190–196 (2003).
    • 12  Woetmann A, Nielsen M, Christensen ST et al. Inhibition of protein phosphatase 2A induces serine/threonine phosphorylation, subcellular redistribution, and functional inhibition of STAT3. Proc. Natl Acad. Sci.96,10620–10625 (1999).
    • 13  Tanaka T, Soriano MA, Grusby MJ. SLIM is a nuclear ubiquitin E3 ligase that negatively regulates STAT signaling. Immunity22,729–736 (2005).
    • 14  Wegenka UM, Buschmann J, Lutticken C, Heinrich PC, Horn F. Acute-phase response factor, a nuclear factor binding to acute-phase response elements, is rapidly activated by interleukin-6 at the post translational level. Mol. Cell. Biol.13,276–288 (1993).
    • 15  Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signaling through the gp130/Jak/STAT pathway. Biochem. J.334,297–314 (1998).
    • 16  Hirano T, Ishihara K, Hibi M. Roles of STAT3 in mediating the cell growth, differentiation and survival signals relayed through the IL-6 family of cytokine receptors. Oncogene19,2548–2556 (2000).
    • 17  Zhang Z, Fuller GM. The competitive binding of STAT3 and NF-κB on an overlapping DNA binding site. Biochem. Biophys. Res. Commun.237,90–94 (1997).
    • 18  Takeda K, Clausen BE, Kaisho T et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of STAT3 in macrophages and neutrophils. Immunity10,39–49 (1999).
    • 19  Pardoll DM. Does the immune system see tumors as foreign or self? Annu. Rev. Immunol.21,807–839 (2003).
    • 20  Wang T, Niu G, Kortylewski M et al. Regulation of the innate and adaptive immune responses by STAT3 signaling in tumor cells. Nature Med.10,48–54 (2004).
    • 21  Cheng F, Wang HW, Cuenca A et al. Critical role for STAT3 signaling in immune tolerance. Immunity19,425–436 (2003).
    • 22  Grandis JR, Drenning SD, Zeng Q et al. Constitutive activation of STAT3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc. Natl Acad. Sci.97,4227–4232 (2000).
    • 23  Niu G, Bowman T, Huang M et al. Roles of activated Src and STAT3 signaling in melanoma tumor cell growth. Oncogene21,7001–7010 (2002).
    • 24  Catlett-Falcone R, Landowski TH, Oshiro MM et al. Constitutive activation of STAT3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity10,105–115 (1999).
    • 25  Epling-Burnette PK, Liu JH, Catlett-Falcone R et al. Inhibition of STAT3 signaling leads to apoptosis of leukemic large granular lymphocytes and decreased Mcl-1 expression. J. Clin. Invest.107,351–362 (2001).
    • 26  Aoki Y, Feldman GM, Tosato G. Inhibition of STAT3 signaling induces apoptosis and decreases survivin expression in primary effusion lymphoma. Blood101,1535–1542 (2003).
    • 27  Shen Y, Devgan G, Darnell JE Jr, Bromberg JF. Constitutively activated STAT3 protects fibroblasts from serum withdrawal and UV-induced apoptosis and antagonizes the proapoptotic effects of activated STAT1. Proc. Natl Acad. Sci.98,1543–1548 (2001).
    • 28  Prendergast GC. Mechanisms of apoptosis by c-Myc. Oncogene18,2967–2987 (1999).
    • 29  Bowman T, Broome MA, Sinibaldi D et al. STAT3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc. Natl Acad. Sci.98,7319–7324 (2001).
    • 30  Buettner R, Mora LB, Jove R. Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention. Clin. Cancer Res.8,945–954 (2002).
    • 31  Kijima T, Niwa H, Steinman RA et al. STAT3 activation abrogates growth factor dependence and contributes to head and neck squamous cell carcinoma tumor growth in vivo. Cell Growth Differ.13,355–362 (2002).
    • 32  Masuda M, Suzui M, Yasumatu R et al. Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res.62,3351–3355 (2002).
    • 33  Sinibaldi D, Wharton W, Turkson J, Bowman T, Pledger WJ, Jove R. Induction of p21WAF1/CIP1 and cyclin D1 expression by the Src oncoprotein in mouse fibroblasts: role of activated STAT3 signaling. Oncogene19,5419–5427 (2000).
    • 34  Vogelstein B, Lane D, Levine AJ. The p53 tumor-suppressor gene integrates numerous signals that control cell life and death. As when a highly connected node in the Internet breaks down, the disruption of p53 has severe consequences. Nature408,307–310 (2000).
    • 35  Mora LB, Buettner R, Seigne J et al. Constitutive activation of STAT3 in human prostate tumors and cell lines direct inhibition of STAT3 signaling induces apoptosis of prostate cancer cells. Cancer Res.62,6659–6666 (2002).
    • 36  Bowman T, Garcia R, Turkson J, Jove R. STATs in oncogenesis. Oncogene19,2474–2488 (2000).
    • 37  Iliopoulos D, Jaeger SA, Hirsch HA, Bulyk ML, Struhl K. STAT3 activation of miR-21 and miR-181b, via PTEN and CYLD, are part of the epigenetic switch linking inflammation to cancer. Mol. Cell39,493–506 (2010).
    • 38  Hanahan D, Weinberg RA. The hallmarks of cancer. Cell100,57–70 (2000).
    • 39  Rak J, Yu JL, Klement G, Kerbel RS. Oncogenes and angiogenesis: signaling three-dimensional tumor growth. J. Invest. Derm.5,24–33 (2000).
    • 40  Grunstein J, Roberts WG, Mathieu-Costello O, Hanahan D, Johnson RS. Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function. Cancer Res.59,1592–1598 (1999).
    • 41  Veikkola T, Karkkainen M, Claesson-Welsh L, Alitalo K. Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Res.60,203–212 (2000).
    • 42  Niu G, Wright KL, Huang M et al. Constitutive STAT3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene21,2000–2008 (2002).
    • 43  Wei D, Le X, Zheng L et al. STAT3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene22,319–329 (2003).
    • 44  Li WC, Ye SL, Sun RX et al. Inhibition of growth and metastasis of human hepatocellular carcinoma by antisense oligonucleotide targeting signal transducer and activator of transcription 3. Clin. Cancer Res.12,7140–7148 (2006).
    • 45  Niu G, Heller R, Catlett-Falcone R et al. Gene therapy with dominant-negative STAT3 suppresses growth of the murine melanoma B16 tumor in vivo. Cancer Res.59,5059–5063 (1999).▪ First validation of STAT3 as a target for cancer therapy in an animal model.
    • 46  Bromberg JF, Wrzeszczynska MH, Devgan G et al. STAT3 as an oncogene. Cell98,295–303 (1999).
    • 47  Siddiquee KAZ, Turkson J. STAT3 as a target for inducing apoptosis in solid and hematological tumors. Cell Res.18(2),254–267 (2008).
    • 48  Turkson J, Jove R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene19(56),6613–6626 (2000).
    • 49  Niu G, Shain K, Huang M et al. Overexpression of a dominant-negative signal transducer and activator of transcription 3 variant in tumor cells leads to production of soluble factors that induce apoptosis and cell cycle arrest. Cancer Res.61,3276–3280 (2001).
    • 50  Turkson J, Ryan D, Kim JS et al. Phosphotyrosyl peptides block STAT3-mediated DNA binding activity, gene regulation, and cell transformation. J. Biol. Chem.276,45443–45455 (2001).▪ Significant proof that small peptide molecules can block cell transformation through a disruption of STAT3 dimerization.
    • 51  Levy DE, Darnell JE Jr. STATs: transcriptional control and biological impact. Nature Rev.3,651–662 (2002).
    • 52  Akira S. Roles of STAT3 defined by tissue-specific gene targeting. Oncogene19,2607–2611 (2000).
    • 53  Turkson J, Zhang S, Palmer J et al. Inhibition of constitutive signal transducer and activator of transcription 3 activation by novel platinum complexes with potent anti-tumor activity. Mol. Cancer Ther.3,1533–1542 (2004).
    • 54  Blaskovich MA, Sun J, Cantor A, Turkson J, Jove R, Sebti SM. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent anti-tumor activity against human and murine cancer cells in mice. Cancer Res.63,1270–1279 (2003).
    • 55  Kortylewski M, Kujawski M, Wang T et al. Inhibiting STAT3 signaling in the hematopoietic system elicits multicomponent anti-tumor immunity. Nat. Med.11,1314–1321 (2005).
    • 56  Catlett-Falcone R, Dalton W, Jove R. STAT proteins as novel target for cancer theraphy. Curr. Opin. Oncol.11(6),490–496 (1999).
    • 57  Costantino L, Barlocco D. STAT 3 as a target for cancer drug discovery. Curr. Med. Chem.15,834–843 (2008).▪▪ Earlier review on the state-of-the-art of STAT3 inhibitors.
    • 58  Yue P, Turkson J. Targeting STAT3 in cancer: how successful are we? Expert Opin. Invest. Drugs18(1),45–56 (2009).
    • 59  Becker S, Groner B, Müller CW. Three-dimensional structure of the STAT3b homodimer bound to DNA. Nature394,145–151 (1998).
    • 60  Fletcher S, Turkson J, Gunning PT. Molecular approches towards the inhibition of the signal transducers and activator of transcription 3 (STAT3) protein. ChemMedChem8,1159–1168 (2008).
    • 61  Ren Z, Cabell LA, Schaefer TS, McMurray JS. Identification of a high-affinity phosphopeptide inhibitor of STAT3. Bioorg. Med. Chem. Lett.13,633–636 (2003).
    • 62  Timofeeva OA, Gaponenko V, Lockett SJ et al. Rationally designed inhibitors identify STAT3 N-domain as a promising anticancer drug target. ACS Chem. Biol.2(12),799–809 (2007).
    • 63  Derossi D, Joliot AH, Chassaing G, Prochiantz A. The third helix of the Antennapedia homeodomain translocates through biological membrane. J. Biol. Chem.269,10444–10450 (1994).
    • 64  Borghouts C, Kunz C, Delis N, Groner D. Monomeric recombinant peptide aptamers are required for efficient intracellular uptake and target inhibition. Mol. Cancer Res.6(2),267–281 (2008).
    • 65  Dourlat J, Liu WQ, Sancier F et al. A novel nonphosphorylated potential anti-tumoral peptide inhibits STAT3 biological activity. Biochimie91,996–1002 (2009).
    • 66  Shuai K, Horvath CM, Huang LH, Qureshi SA, Cowburn D, Darnell JE. Interferon activation of the transcription factor STAT91 involves dimerization through SH2–phosphotyrosyl peptide interactions. Cell76(5),821–828 (1994).
    • 67  Vultur A, Cao J, Arulanandam R et al. Cell-to-cell adhesion modulates STAT3 activity in normal and breast carcinoma cells. Oncogene2315,2600–2616 (2004).
    • 68  Ren Z, Coleman DR, Mandal PK et al. Peptidomimetic inhibitors of STAT3: structure–activity relationships and cellular activity. Presented at: The 16th EORTC-NCI-AACR, Symposium on Molecular Targets and Cancer Therapeutics. Genève, Switzerland, 28 September–1 October 2004.
    • 69  Shuai K. Regulation of cytokine signaling pathways by PIAS proteins. Cell Res.16,196–202 (2006).
    • 70  Borghouts C, Tittmann H, Delis N, Kirchenbauer M, Brill B, Groner B. The intracellular delivery of a recombinant peptide derived from the acidic domain of PIAS3 inhibits STAT3 transactivation and induces tumor cell death. Mol. Cancer Res.8(4),539–553 (2010).
    • 71  Turkson J, Kim JS, Zhang S et al. Novel peptidomimetic inhibitors of signal transducers and activator of transcription 3 dimerization and biological activity. Mol. Cancer Ther.3,261–269 (2004).
    • 72  Gunning PT, Katt WP, Glenn M et al. Isoform selective inhibition of STAT1 or STAT3 homo-dimerization via peptidomimetic probes: structural recognition of STAT SH2 domains. Bioorg. Med. Chem. Lett.17,1875–1878 (2007).
    • 73  McMurray JS. Structural basis for the binding of high affinity phosphopeptides to STAT3. Biopolymers Peptide90(1),69–79 (2007).
    • 74  Schust J, Berg T. A high-throughput fluorescence polarization assay for signal transducers and activator of transcription 3. Anal. Biochem.330,114–118 (2004).
    • 75  Mandal PK, Heard PA, Ren Z, Chen X, McMurray JS. Solid-phase synthesis of STAT3 inhibitors incorporating O-carbamoylserine and O-carbamoylthreonine as glutamine mimics. Bioorg. Med. Chem. Lett.17,654–656 (2007).
    • 76  Mandal PK, Ren Z, Chen X, Xiong C, McMurray JS. Structure–affinity relationships of glutamine mimics incorporated into phosphopeptides targeted to the SH2 domain of signal transducer and activator of transcription 3. J. Med. Chem.52,6126–6141 (2009).
    • 77  Coleman DR, Kaluarachichi K, Ren Z, Chen X, McMurray JS. Solid-phase synthesis of phosphopeptides incorporating 2,2-dimethyloxazolidine pseudoproline analogues: evidence for trans Leu-Pro peptide bonds in STAT3 inhibitors. Int. J. Pept. Res. Ther.14(1),1–9 (2008).
    • 78  Chen J, Nikolovska-Coleska Z, Yang CY et al. Design and synthesis of a new, conformationally constrained, macrocyclic small-molecule inhibitor of STAT3 via click chemistry. Bioorg. Med. Chem. Lett.17,3939–3942 (2007).
    • 79  Dourlat J, Valentin B, Liu WQ, Garbay C. New synthesis of tetrazolylmethylphenylalanine and O-malonyltyrosine as pTyr mimetics for the design of STAT3 dimerization inhibitors. Bioorg. Med. Chem. Lett.17,3943–3946 (2007).
    • 80  Gomez C, Bai L, Zhang J et al. Design, synthesis, and evalutation of peptidomimetics containing Freidinger lactams as STAT3 inhibitors. Bioorg. Med. Chem. Lett.19,1733–1736 (2009).
    • 81  Coleman DR, Ren Z, Mandal PK et al. Investigation of the binding determinants of phosphopeptides targeted to the SRC homology 2 domain of the signal transducer and activator of transcription 3. Development of a high-affinity peptide inhibitor. J. Med. Chem.48,6661–6670 (2005).
    • 82  Mandal PK, Liao WSL, McMurray JS. Synthesis of phosphatase-stable, cell-permeable peptidomimetic prodrugs that target the SH2 domain of STAT3. Org. Lett.11(15),3394–3397 (2009).
    • 83  Chen J, Bai L, Bernard D et al. Structure-based design of conformationally constrained, cell-permeable STAT3 inhibitors. ACS Med. Chem. Lett.1(2),85–89 (2010).
    • 84  Weidler M, Rether J, Anke T, Erkel G. Inhibition of interleukin-6 signaling by galiellalactone. FEBS Lett.484,1–6 (2000).
    • 85  Shin DS, Kim HN, Shin KD et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res.69(1),193–202 (2009).
    • 86  Siddiquee K, Gunning PT, Glenn M et al. An oxazole-based small-molecule STAT3 inhibitor modulates STAT3 stability and processing and induces anti-tumor cell effects. ACS Chem. Biol.2(12),787–798 (2007).
    • 87  Fletcher S, Singh J, Zhang X et al. Disruption of transcriptionally active STAT3 dimers with nonphosphorylated, salicylic acid-based small molecules: potent in vitro and tumor cell activities. ChemBioChem10(12),1959–1964 (2009).
    • 88  Gunning PT, Glenn MP, Siddiquee K et al. Targeting protein–protein interactions: suppression of STAT3 dimerization with rationally designed small-molecule, nonpeptidic SH2 domain binders. Chem. Bio. Chem.9(17),2800–2803 (2008).
    • 89  Ge J, Wu H, Yao SQ. An unnatural amino acid that mimics phosphotyrosine. Chem. Commun. (Camb.)46(17),2980–2982 (2010).
    • 90  Siddiquee K, Zhang S, Guida WC et al. Selective chemical probe inhibitor of STAT3, identified through structure-based virtual screening, induces anti-tumor activity. Proc. Natl Acad. Sci. USA104(18),7391–7396 (2007).
    • 91  Zhang X, Yue P, Fletcher S, Zhao W, Gunning PT, Turkson J. A novel small-molecule disrupts STAT3 SH2 domain–phosphotyrosine interactions and STAT3-dependent tumor processes. Biochem. Pharmacol.79(10),1398–1409 (2010).
    • 92  Song H, Wang R, Wang S, Lin J. A low-molecular-weight compound discovered through virtual database screening inhibits STAT3 function in breast cancer cells. Proc. Natl Acad. Sci. USA102(13),4700–4705 (2005).
    • 93  Fuh B. Sobo M, Cen L et al. LLL-3 inhibits STAT3 activity, suppresses glioblastoma cell growth and prolongs survival in a mouse glioblastoma model. Br. J. Cancer100(1),106–112 (2009).
    • 94  Bhasin D, Cisek K, Pandharkar T et al. Design, synthesis, and studies of small molecule STAT3 inhibitors. Bioor. Med. Chem. Lett.18(1),391–395 (2008).
    • 95  Lin L, Brian H, Pui-Kai L et al. A novel small molecule, LLL12 inhibits STAT3 phosphorylation and activities and exhibits potent growth-suppressive activity in human cancer cells. Neoplasia12(1),39–50 (2010).
    • 96  Cui JR, Jian YJ, Wu YK, Wang SM. Synthesis of potential inhibitors for STAT-3 dimerization. Chinese J. Chem.24,1163–1169 (2006).
    • 97  Uehara Y, Mochizuki M, Matsuno K, Haino T, Asai A. Novel high-throughput screening system for identifying STAT3-SH2 antagonists. Biochem. Biophys. Res. Comm.380(3),627–631 (2009).
    • 98  Hao W, Hu Y, Niu C et al. Discovery of catechol structural moiety as a STAT3 SH2 domain inhibitor by virtual screening. Bioorg. Chem. Lett.18,4988–4992 (2008).
    • 99  Shahani VM, Yue P, Haftchenary S et al. Identification of purine-scaffold small-molecule inhibitors of STAT3 activation by QSAR studies. ACS Med. Chem. Lett.2(1),79–84 (2011).
    • 100  Turkson J, Zhang S, Palmer J et al. Inhibition of constitutive signal transducer and activator of transcription 3 activation by novel platinum complexes with potent anti-tumor activity. Mol. Cancer Ther.3(12),1533–1542 (2004).
    • 101  Turkson J, Zhang S, Mora LB et al. A novel platinum compound inhibits constitutive STAT3 signaling and induces cell cycle arrest and apoptosis of malignant cells. J. Biol. Chem.280(38),32979–32988 (2005).
    • 102  Xu X, Kasembeli MM, Jiang X, Tweardy BJ, Tweardy DJ. Chemical probes that competitively and selectively inhibit STAT3 activation. PLoS One4(3),e4783 (2009).
    • 103  Matsuno K, Masuda Y, Uehara Y et al. Identification of a new series of STAT3 inhibitors by virtual screening. ACS Med. Chem. Lett.1(8),371–375 (2010).
    • 104  Williamson JR. G-quartet structures in telomeric DNA. Annu. Rev. Biophys. Biomol. Struct.23,703–730 (1994).
    • 105  Burge S, Parkinson GN, Hazel P, Todd AK, Neidle S. Quadruplex DNA: sequence, topology and structure. Nucleic Acids Res.34(19),5402–5415 (2006).
    • 106  Jing N, Zhu Q, Yuan P, Li Y, Mao L, Tweardy DJ. Targeting signal transducer and activator of transcription 3 with G-quartet oligonucleotides: a potential novel therapy for head and neck cancer. Mol. Cancer Ther.5(2),279–286 (2006).
    • 107  Turkson J, Zhang S, Mora LB, Burns A, Sebti S, Jove R. A novel platinum compound inhibits constitutive STAT3 signaling and induces cell cycle arrest and apoptosis of malignant cells. J. Biol. Chem.280(38),32979–32988 (2005).
    • 108  Weerasinghe P, Garcia GE, Zhu Q et al. Inhibition of STAT3 activation and tumor growth suppression of nonsmall cell lung cancer by G-quartet oligonucleotides. Int. J. Onc.31,129–136 (2007).
    • 109  Leong PL, Andrews GA, Johnson DE et al. Targeted inhibition of STAT3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc. Natl Acad. Sci. USA100,4138–4143 (2003).
    • 110  Barton BE, Murphy TF, Shu P, Huang HF, Meyenhofer M, Barton A. Novel single-stranded oligonucleotides that inhibit signal transducer and activator of transcription 3 induce apoptosis in vitro and in vivo in prostate cancer cell lines. Mol. Cancer Ther.3,1183–1191 (2004).
    • 111  Hbibi T, Laguillier C, Souissi I et al. Efficient killing of SW480 colon carcinoma cells by a signal transducer and activator of transcription (STAT) 3 hairpin decoy oligodeoxynucleotide – interference with interferon-c-STAT1-mediated killing. FEBS J.276,2505–2515 (2009).
    • 112  Zhanga X, Liua P, Zhang B, Wanga A, Yang M. Role of STAT3 decoy oligodeoxynucleotides on cell invasion and chemosensitivity in human epithelial ovarian cancer cells. Cancer Genet. Cytogen.197,46–53 (2010).
    • 113  Masuda M, Suzui M, Weinstein B. Effect of epigallocatechin-3-gallate on growth, epidermal growyh factor receptor signalling pathways, gene expression, and chemosensitivity in human head and neck squamous cell carcinoma cell lines. Clin. Cancer Res.7,4220–4229 (2001).
    • 114  Way TD, Lee JC, Kuo DH et al. Inhibition of epidermal growth factor receptor signaling by Saussurea involucrata, a rare traditional Chinese medicinal herb, in human hormone-resistant prostate cancer PC-3 cells. J. Agric. Food Chem.58(6),3356–3365 (2010).
    • 115  Zhou J, Ong C-N, Hur G-M, Shen H-M. Inhibition of the JAK-STAT3 pathway by andrographolide enhances chemosensitivity of cancer cells to doxorubicin. Biochem. Pharmacol.79,1242–1250 (2010).
    • 116  Wesolowska O, Kuźdźal M, Štrancar T, Michalak K. Interaction of the chemopreventive agent resveratrol and its metabolite, piceatannol, with model membranes. Biochim. Biophys. Acta1788,1851–1860 (2009).
    • 117  Su L, David M. Distinct mechanisms of STAT phosphorylation via the interferon-α/β receptor. Selective inhibition of STAT3 and STAT5 by piceatannol. J. Biol. Chem.275,12661–12666 (2000).
    • 118  Sandra MLT, Li F, Rajendran P, Kumar AP, Hui KM, Sethi G. Identification of β-escin as a novel inhibitor of STAT3/JAK2 signaling pathway that suppresses proliferation and induces apoptosis in human hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther.334,285–293 (2010).
    • 119  Wang YMX, Yan S, Shen S et al. 17-hydroxy-jolkinolide B inhibits signal transducers and activators of transcription 3 signaling by covalently cross-linking Janus kinases and induces apoptosis of human cancer cells. Cancer Res.69(18),7302–7310 (2009).
    • 120  Sandur SK, Pandey MK, Sung B, Aggarwal BB. 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.8(1),107–118 (2010).
    • 121  Ito S, Oyake T, Murai K, Ishida Y. Deguelin suppresses cell proliferation via the inhibition of survivin expression and STAT3 phosphorylation in HTLV-1-transformed T cells. Leukemia Res.34(3),352–357 (2010).
    • 122  Dixit D, Sharma V, Ghosh S, Koul N, Mishra PK, Sen E. Manumycin inhibits STAT3, telomerase activity, and growth of glioma cells by elevating intracellular reactive oxygen species generation. Free Radic. Biol. Med.47,364–374 (2009).
    • 123  Zhang J, Li L, Kim SH, Hagerman AE, Lu J. Anticancer, antidiabetic and other pharmacologic and biological activities of penta-galloyl-glucose. Pharm. Res.26(9),2066–2080 (2009).
    • 124  Hu H, Lee HJ, Jiang C et al. Penta-1,2,3,4,6-O-galloyl-B-D-glucose induces p53 and inhibits STAT3 in prostate cancer cells in vitro and suppresses prostate xenograft tumor growth in vivo. Mol. Cancer Ther.7(9),2681–2691 (2008).
    • 125  Agarwal C, Tyagi A, Kaur M, Agarwal R. Silibinin inhibits constitutive activation of STAT3, and causes caspase activation and apoptotic death of human prostate carcinoma DU145 cells. Carcinogenesis28,1463–1470 (2007).
    • 126  Sobota R, Szwed M, Kasza A, Bugno M, Kordula T. Parthenolide inhibits activation of signal transducers and activators of trascription (STATs) induced by cytokines of the IL-6 family. Biochem. Biophys. Res. Commun.267,329–333 (2000).
    • 127  Del Bano MJ, Lorente J, Castillo J et al. Phenolic diterpenes, flavones, and rosmarinic acid distribution during the development of leaves, flowers, stems, and roots of Rosmarinus officinalis. Antioxidant activity. J. Agric. Food Chem.51,4247–4253 (2003).
    • 128  Lai CS, Lee HJ, Ho CT et al. Rosmanol potently inhibits lipopolysaccharide-induced iNOS and COX-2 expression through downregulating MAPK, NF-KB, STAT3 and C/EBP signaling pathways. J. Agric. Food Chem.57,10990–10998 (2009).
    • 129  Zhang C, Li B, Gaikwad AS et al. Avicin D selectively induces apoptosis and downregulates p-STAT-3, Bcl-2, and survivin incutaneous T-cell lymphoma cells. J. Invest. Derm.128,2728–2735 (2008).
    • 130  Wang Z, Jin H, Xu R, Mei Q, Fan D. Triptolide downregulates Rac1 and the JAK/STAT3 pathway and inhibits colitis-related colon cancer progression. Exp. Mol. Med.41(10),717–727 (2009).
    • 131  Lin L, Hutzen B. Zuo M et al. Novel STAT3 phosphorylation inhibitors exhibit potent growth-suppressive activity in pancreatic and breast cancer cells. Cancer Res.70(6),2445–2454 (2010).
    • 132  Kim BH, Oh SR, Yin CH et al. MS-1020 is a novel small molecule that selectively inhibits JAK3 activity. Br. J. Haematol.148,132–143 (2009).
    • 133  Kim BH, Yin CH, Guo Q et al. A small-molecule compound identified through a cell-based screening inhibits JAK/STAT pathway signaling in human cancer cells. Mol. Cancer Ther.7(9),2672–2680 (2008).
    • 134  Lasho TL, Tefferi A, Hood JD, Verstovsek S, Gilliland DG, Pardanani A. TG101348, a JAK2-selective antagonist, inhibits primary hematopoietic cells derived from myeloproliferative disorder patients with JAK2V617F, MPLW515K or JAK2 exon 12 mutations as well as mutation negative patients. Leukemia22(9),1790–1792 (2008).
    • 135  Cotto M, Cabanillas F, Tirado M, Garcia MV, Pacheco E. Epigenetic therapy of lymphoma using histone deacetylase inhibitors. Clin. Transl. Oncol.12(6),401–409 (2010).
    • 136  Verstovsek S. Therapeutic potential of JAK2 inhibitors. Hematology1,636–642 (2009).
    • 137  Lin KL, Su JC, Chien CM et al. Naphtho[1,2-b]furan-4,5-dione disrupts Janus kinase-2 and induces apoptosis in breast cancer MDA-MB-231. Toxicol. In Vitro24(4),1158–1167 (2010).
    • 138  Hedvat M, Huszar D, Herrmann A et al. The JAK2 inhibitor AZD1480 potently blocks STAT3 signaling and oncogenesis in solid tumors. Cancer Cell16(6),487–497 (2009).
    • 139  Scuto A, Krejci P, Popplewell L et al. The novel JAK inhibitor AZD1480 blocks STAT3 and FGFR3 signaling, resulting in suppression of human myeloma cell growth and survival. Leukemia DOI: 10.1038/leu.2010.289 (2010) (Epub ahead of print).
    • 140  Ioannidis S, Lamb ML, Wang T et al. Discovery of 5-chloro-N2-[(1S)-1-(5-fluoropyrimidin-2-yl)ethyl]-N4-(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (AZD1480) as a novel inhibitor of the Jak/STAT pathway. J. Med Chem.54(1),262–276 (2011).
    • 141  Verstovsek S, Kantarjian H, Mesa RA et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N. Engl. J. Med.363(12),1117–1127 (2010).
    • 142  Mesa RA. Ruxolitinib, a selective JAK1 and JAK2 inhibitor for the treatment of myeloproliferative neoplasms and psoriasis. Drugs13(6),394–403 (2010).
    • 143  Campas-Moya C. Ruxolitinib: tyrosine-protein kinase JAK2 inhibitor: treatment of myelofibrosis: treatment of myeloproliferative neoplasms: treatment pf psoriasis. Drugs Fut.35(6),457–465 (2010).
    • 144  Pardanani A, Lasho T, Smith G, Burns CJ, Fantino E, Tefferi A. CYT387, a selective JAK1/JAK2 inhibitor: in vitro assessment of kinase selectivity and preclinical studies using cell lines and primary cells from polycythemia vera patients. Leukemia23(8),1441–1445 (2009).
    • 145  Burns CJ, Bourke DG, Andrau L et al. Phenylaminopyrimidines as inhibitors of Janus kinases (JAKs). Bioorg. Med. Chem. Lett.19(20),5887–5892 (2009).
    • 146  Pedranzini L, Dechow T, Berishaj M et al. Pyridone 6, a pan-Janus-activated kinase inhibitor, induces growth inhibition of multiple myeloma cells. Cancer Res.66(19),9714–9721 (2006).
    • 147  Quintás-Cardama A, Manshouri T, Estrov Z et al. Preclinical characterization of atiprimod, a novel JAK2 and JAK3 inhibitor. Invest. New Drugs DOI: 10.1007/s10637-010-9429-z (2010). (Epub ahead of print).
    • 148  Vazina MA, Shishodia D, Harris D et al. Atiprimod blocks STAT3 phosphorilation and induces apoptosis in multiple myeloma cells. Br. J. Cancer93,70–80 (2005).
    • 149  Ramakrishnan V, Timm M, Haug JL et al. Sorafenib, a dual Raf kinase/vascular endothelial growth factor receptor inhibitor has significant antimyeloma activity and synergizes with common antimyeloma drugs. Oncogene29(8),1190–1202 (2010).
    • 150  Yang F, Brown C, Buettner R et al. Sorafenib induces growth arrest and apoptosis of human glioblastoma cells through the dephosphorylation of signal transducers and activators of transcription 3. Mol. Cancer Ther.9(4),953–962 (2010).
    • 151  Sen B, Saigal B, Parikh N, Gallik G, Johnson FM. Sustained Src inhibition results in signal transducer and activator of transcription 3 (STAT3) activation and cancer cell survival via altered Janus-activated kinase-STAT3 binding. Cancer Res.69(5),1958–1965 (2009).
    • 152  Averett Byers L, Sen B, Saigal B et al. Reciprocal regulation of c-Srg and STAT3 in nonsmall cell lung cancer. Clin. Cancer Res.12(22),6852–6861 (2009).
    • 153  Premkumar DR, Jane EP, Agostino NR, Scialabbar JL, Pollack IF. Dasatinib synergizes with JSI-124 to inhibit growth and migration and induce apoptosis of malignant human glioma cells. J. Carcinog.9(7),1–14 (2010).
    • 154  Hutzen B, Friedman L, Sobo M et al. Curcumin analogue GO-Y030 inhibits STAT3 activity and cell growth in breast and pancreatic carcinomas. Int. J. Oncol.35(4),867–872 (2009).
    • 155  Keinosuke R, Michiro S, Edwin C et al. Oleanane triterpenoid CDDO-Me induces apoptosis in multidrug resistant osteosarcoma cells through inhibition of STAT3 pathway. BMC Cancer10,187–197 (2010).
    • 156  Dowlati A, Kluge A, Nethery D, Halmos B, Kern J. SCH66336, inhibitor of protein farnesylation, blocks signal transducer and activators of transcription 3 signaling in lung cancer and interacts with a small molecule inhibitor of epidermal growth factor receptor/human epidermal growth factor receptor 2. AntiCancer Drugs19(1),9–16 (2008).
    • 157  Venkatasubbarao K, Choudary A, Freeman JW. Farnesyl transferase inhibitor (R115777)-induced inhibition of STAT3 (Tyr705) phosphorylation in human pancreatic cancer cell lines require extracellular signal-regulated kinases. Cancer Res.65(7),2861–2871 (2005).
    • 158  Horiguchi A, Asano T, Kuroda K et al. STAT3 inhibitor WP1066 as a novel therapeutic agent for renal cell carcinoma. Br. J. Cancer102(11),1592–1599 (2010).
    • 159  Ren X, Duan L, He Q et al. Identification of niclosamide as a new small-molecule inhibitor of the STAT3 signaling pathway. ACS Med. Chem. Lett.1(9),454–459 (2010).
    • 160  Bhargava A, Vaishampayan UN. Satraplatin: leading the new generation of oral platinum agents. Expert Opin. Investig. Drugs18(11),1787–1797 (2009).
    • 161  Gariboldi MB, Ravizza R, Molteni R, Osella D, Gabano E, Monti E. Inhibition of STAT3 increases doxorubicin sensitivity in human metastatic breast cancer cell line. Cancer Lett.258,181–188 (2007).
    • 162  Ngan CY, Yamamoto H, Takagi A et al. Oxaliplatin induces mitotic catastrophe and apoptosis in esophageal cancer cells. Cancer Sci.99(1),129–139 (2008).
    • 163  Shin D-S, Masciocchi D, Gelain A et al. Synthesis, modeling, and crystallographic study of 3,4-disubstituted-1,2,5-oxadiazoles and evaluation of their ability to decrease STAT3 activity. Med. Chem. Comm.1(2),156–164 (2010).
    • 164  Jougasaki M, Ichiki T, Takenoshita Y, Setoguchi M. Statins suppress interleukin-6-induced monocyte chemo-attractant protein-1 by inhibiting Janus kinase/signal transducers and activators of transcription pathways in human vascular endothelial cells. Br. J. Pharmacol.159(6),1294–1303 (2010).
    • 165  Lee CS, Shin YJ, Won C et al. Simvastatin acts as an inhibitor of interferon γ-induced cycloxygenase-2 expression in human THP-1 cells, but not in murine RAW264.7 cells. Biocell.33(2),107–114 (2009).
    • 166  Jung JE, Kim HS, Lee CS et al. Caffeic acid and its synthetic derivative CADPE suppress tumor angiogenesis by blocking STAT3-mediated VEGF expression in human renal carcinoma cells. Carcinogenesis28(8),1780–1787 (2007).
    • 167  Masahiro O, Naoko S, Masanobu S et al. 12-O-tetradecanoylphorbol-13-acetate inhibits melanoma growth by inactivation of STAT3 through protein kinase c-activated tyrosine phosphatase(s). J. Biol. Chem.284(44),30416–30423 (2009).
    • 168  Yiin JJ, Hu B, Schornack PA et al. ZD6474, a multitargeted inhibitor for receptor tyrosine kinases, suppresses growth of gliomas expressing an epidermal growth factor receptor mutant, EGFRvIII, in the brain. Mol. Cancer Ther.9(4),929–941 (2010).
    • 169  Uckun FM, Dibirdik I, Qazi S, Yiv S. Therapeutic nanoparticle constructs of a JAK3 tyrosine kinase inhibitor against human B-lineage ALL cells. Arzneimittelforschung Journal60(4),210–217 (2010).
    • 170  Gariboldi MB, Ravizza R, Monti E. The IGFR1 inhibitor NVP-AEW541 disrupts a pro-survival and pro-angiogenic IGF-STAT3-HIF1 pathway in human glioblastoma cells. Biochem. Pharmacol.80(4),455–462 (2010).
    • 171  Yang Y, Takayuki I, Tamotsu T et al. HIV-1 protease inhibitor induces growth arrest and apoptosis of human prostate cancer LNCaP cells in vitro and in vivo in conjunction with blockade of androgen receptor STAT3 and AKT signaling. Cancer Science96(7),425–433 (2005).
    • 172  Nelson EA, Walker SR, Kepich A et al. Nifuroxazide inhibits survival of multiple myeloma cells by directly inhibiting STAT3. Blood112(13),5095–5102 (2008).
    • 173  Kim HW, Kim JL, Lee HK, Hur DY, Yun IH, Kim SD. Enalapril alters expression of key growth factors in experimental diabetic retinopathy. Curr. Eye Res.34(11),976–987 (2009).
    • 174  Yu ZY, Huang R, Xiao H et al. Fluacrypyrim, a novel STAT3 activation inhibitor, induces cell cycle arrest and apoptosis in cancer cells harboring constitutively-active STAT3. Int. J. Cancer127(6),1259–1270 (2010).
    • 175  Shao W, Growney JD, Feng Y et al. Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: defining molecular mechanisms of resistance. Int. J. Cancer127(9),2199–2208 (2010).
    • 176  Changyou L, Yan Z, Malabika S et al. Bortezomib up-regulates activated signal transducer and activator of transcription-3 and synergizes with inhibitors of signal transducer and activator of transcription-3 to promote head and neck squamous cell carcinoma cell death. Mol. Cancer Ther.8(8),2211–2220 (2009).
    • 177  Korehito K, Nantiga V, Kayono H et al. A redox-silent analogue of tocotrienol acts as a potential cytotoxic agent against human mesothelioma cells. Life Sciences84(19–20),650–656 (2009).
    • 178  Lau CK, Yang ZF, Lam SP et al. Inhibition of STAT3 activity by YC-1 enhances chemo-sensitivity in hepatocellular carcinoma. Cancer Biol. Ther.6(12),1900–1907 (2007).
    • 179  Selvendiran K, Bratasz A, Tong L, Ignarro LJ, Kuppusamy P. NCX-4016, a nitro-derivative of aspirin, inhibits EGFR and STAT3 signaling and modulates Bcl-2 proteins in cisplatin-resistant human ovarian cancer cells and xenografts. Cell Cycle7(1),81–88 (2008).
    • 180  Chatterjee PK, Al-Abed Y, Sherry B, Metz CN. Cholinergic agonists regulate JAK2/STAT3 signaling to suppress endothelial cell activation. Am. J. Physiol. Cell Physiol.297(5),C1294–C1306 (2009).
    • 181  Lee HS, Han J, Lee SH, Park JA, Kim KW. Meteorin promotes the formation of GFAP-positive glia via activation of the Jak-STAT3 pathway. J. Cell Sci.123(11),1959–1968 (2010).
    • 182  Nakaya A, Sagawa M, Muto A, Uchida H, Ikeda Y, Kizaki M. The gold compound auranofin induces apoptosis of human multiple myeloma cells through both down-regulation of STAT3 and inhibition of NF-κB activity. Leuk. Res.35(2),243–249 (2010).
    • 183  McCleese JK, Bear MD, Fossey SL et al. The novel HSP90 inhibitor STA-1474 exhibits biologic activity against osteosarcoma cell lines. Int. J. Cancer125(12),2792–2801 (2009).
    • 201  Government of the USA. WO 2008151037 (2008).
    • 202  Orchid Research Laboratories Limited. WO 2007042912 (2007).
    • 203  Baylor College Medicine. WO 2006069001 (2006).
    • 204  University of Central Florida Research Foundation, Inc. WO 2010033685 (2010).
    • 205  H. Lee Moffitt Cancer Center & Research Institute. WO 2007136858 (2007).
    • 206  Farjo R. WO 2009042677 (2009).
    • 207  Baylor College Medicine. WO 2009149192 (2009).
    • 208  Pharma IP General Incorporate Association. WO 2010004761 (2010).
    • 209  Univesity of South Florida. WO 2007US2827 (2007).
    • 210  Supergen, Inc. WO 2010011349 (2010).
    • 211  Orchid Research Laboratories Limited. WO 2007042912 (2007).
    • 212  Orchid Research Laboratories Limited. WO 2009060282 (2009).
    • 213  Boston Biomedical, Inc. WO 2009036059 (2009).
    • 214  Otsuka Pharmaceutical Co, Ltd. WO 2009057811 (2009).
    • 215  Board of Regents, the Univesity of Texas System. WO 2010005807 (2010).
    • 216  Kwon BM. US 20080051442 (2008).