The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Structure of STATs
STAT Alternative Isoforms
3. STAT Activation and Signaling
4. The Effect of STATs on OvCa Cells
4.1. STAT1
4.2. STAT2
4.3. STAT3
4.3.1. Proliferation and Survival
4.3.2. Angiogenesis
4.3.3. Metastasis
4.3.4. Stemness
4.4. STAT4
4.5. STAT5
4.6. STAT6
5. The Role of STATs in the TME
5.1. Macrophages
5.2. Dendritic Cells
5.3. MDSC
5.4. Tregs
6. STAT Inhibitors
6.1. STAT1
6.2. STAT3
6.2.1. Natural Compounds
6.2.2. Synthetic Inhibitors
6.3. STAT5
6.4. STAT6
7. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Asare-Werehene, M.; Tsuyoshi, H.; Zhang, H.; Salehi, R.; Chang, C.-Y.; Carmona, E.; Librach, C.L.; Mes-Masson, A.-M.; Chang, C.-C.; Burger, D.; et al. Plasma Gelsolin Confers Chemoresistance in Ovarian Cancer by Resetting the Relative Abundance and Function of Macrophage Subtypes. Cancers 2022, 14, 1039. [Google Scholar] [CrossRef]
- Baldwin, L.A.; Huang, B.; Miller, R.W.; Tucker, T.; Goodrich, S.T.; Podzielinski, I.; DeSimone, C.P.; Ueland, F.R.; Van Nagell, J.R.; Seamon, L.G. Ten-year relative survival for epithelial ovarian cancer. Obstet. Gynecol. 2012, 120, 612–618. [Google Scholar] [CrossRef] [PubMed]
- Modugno, F.; Edwards, R.P. Ovarian cancer: Prevention, detection, and treatment of the disease and its recurrence. molecular mechanisms and personalized medicine meeting report. Int. J. Gynecol. Cancer 2012, 22, S45–S57. [Google Scholar] [CrossRef] [PubMed]
- Colomiere, M.; Ward, A.C.; Riley, C.; Trenerry, M.K.; Cameron-Smith, D.; Findlay, J.; Ackland, L.; Ahmed, N. Cross talk of signals between EGFR and IL-6R through JAK2/STAT3 mediate epithelial-mesenchymal transition in ovarian carcinomas. Br. J. Cancer 2009, 100, 134–144. [Google Scholar] [CrossRef]
- Huang, M.; Page, C.; Reynolds, R.; Lin, J. Constitutive activation of stat 3 oncogene product in human ovarian carcinoma cells. Gynecol. Oncol. 2000, 79, 67–73. [Google Scholar] [CrossRef] [PubMed]
- Abubaker, K.; Luwor, R.B.; Zhu, H.; McNally, O.; Quinn, M.A.; Burns, C.J.; Thompson, E.W.; Findlay, J.K.; Ahmed, N. Inhibition of the JAK2/STAT3 pathway in ovarian cancer results in the loss of cancer stem cell-like characteristics and a reduced tumor burden. BMC Cancer 2014, 14, 317. [Google Scholar] [CrossRef]
- Wang, Z.; Chen, W.; Zuo, L.; Xu, M.; Wu, Y.; Huang, J.; Zhang, X.; Li, Y.; Wang, J.; Chen, J.; et al. The Fibrillin-1/VEGFR2/STAT2 signaling axis promotes chemoresistance via modulating glycolysis and angiogenesis in ovarian cancer organoids and cells. Cancer Commun. 2022, 42, 245–265. [Google Scholar] [CrossRef] [PubMed]
- Mowery, A.S.; Klockowski, R.J.; Davis, E. Simultaneous versus independent polymerization of a bonding agent and composite resin. Oper. Dent. 1986, 11, 69–73. [Google Scholar]
- Ihle, J.N. STATs: Signal Transducers and activators of transcription. Cell 1996, 84, 331–334. [Google Scholar] [CrossRef]
- Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
- Ivashkiv, L.B.; Hu, X. Signaling by STATs. Arthritis Res. Ther. 2004, 6, 159–168. [Google Scholar] [CrossRef] [PubMed]
- Khalaf, K.; Hana, D.; Chou, J.T.-T.; Singh, C.; Mackiewicz, A.; Kaczmarek, M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front. Immunol. 2021, 12, 656364. [Google Scholar] [CrossRef] [PubMed]
- Klemm, F.; Joyce, J.A. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015, 25, 198–213. [Google Scholar] [CrossRef] [PubMed]
- Gao, S.; Zhang, W.; Yan, N.; Li, M.; Mu, X.; Yin, H.; Wang, J. The impact of STAT3 and phospho-STAT3 expression on the prognosis and clinicopathology of ovarian cancer: A systematic review and meta-analysis. J. Ovarian Res. 2021, 14, 164. [Google Scholar] [CrossRef]
- Ying, X.; Wu, Q.; Wu, X.; Zhu, Q.; Wang, X.; Jiang, L.; Chen, X.; Wang, X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 2016, 7, 43076–43087. [Google Scholar] [CrossRef]
- Yu, T.; Gan, S.; Zhu, Q.; Dai, D.; Li, N.; Wang, H.; Chen, X.; Hou, D.; Wang, Y.; Pan, Q.; et al. Modulation of M2 macrophage polarization by the crosstalk between Stat6 and Trim24. Nat. Commun. 2019, 10, 4353. [Google Scholar] [CrossRef]
- Zorn, E.; Nelson, E.A.; Mohseni, M.; Porcheray, F.; Kim, H.; Litsa, D.; Bellucci, R.; Raderschall, E.; Canning, C.; Soiffer, R.J.; et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006, 108, 1571–1579. [Google Scholar] [CrossRef]
- Chen, W.; Jin, W.; Hardegen, N.; Lei, K.-J.; Li, L.; Marinos, N.; McGrady, G.; Wahl, S.M. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 2003, 198, 1875–1886. [Google Scholar] [CrossRef]
- McHugh, M.D.; Park, J.; Uhrich, R.; Gao, W.; Horwitz, D.A.; Fahmy, T.M. Paracrine co-delivery of TGF-beta and IL-2 using CD4-targeted nanoparticles for induction and maintenance of regulatory T cells. Biomaterials 2015, 59, 172–181. [Google Scholar] [CrossRef]
- Chen, Q.; Kim, Y.C.; Laurence, A.; Punkosdy, G.A.; Shevach, E.M. IL-2 controls the stability of Foxp3 expression in TGF-β–induced Foxp3+ T cells in vivo. J. Immunol. 2011, 186, 6329–6337. [Google Scholar] [CrossRef]
- Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
- Perrone, G.; Ruffini, P.A.; Catalano, V.; Spino, C.; Santini, D.; Muretto, P.; Spoto, C.; Zingaretti, C.; Sisti, V.; Alessandroni, P.; et al. Intratumoural FOXP3-positive regulatory T cells are associated with adverse prognosis in radically resected gastric cancer. Eur. J. Cancer 2008, 44, 1875–1882. [Google Scholar] [CrossRef] [PubMed]
- El Andaloussi, A.; Lesniak, M.S. CD4+ CD25+ FoxP3+ T-cell infiltration and heme oxygenase-1 expression correlate with tumor grade in human gliomas. J. Neuro Oncol. 2007, 83, 145–152. [Google Scholar] [CrossRef] [PubMed]
- Hendry, L.; John, S. Regulation of STAT signalling by proteolytic processing. JBIC J. Biol. Inorg. Chem. 2004, 271, 4613–4620. [Google Scholar] [CrossRef]
- Lim, C.P.; Cao, X. Structure, function, and regulation of STAT proteins. Mol. Biosyst. 2006, 2, 536–550. [Google Scholar] [CrossRef] [PubMed]
- Pellegrini, S.; Dusanter-Fourt, I. The structure, regulation and function of the Janus kinases (JAKs) and the signal transducers and activators of transcription (STATs). JBIC J. Biol. Inorg. Chem. 1997, 248, 615–633. [Google Scholar] [CrossRef]
- Heinrich, P.C.; Behrmann, I.; Müller-Newen, G.; Schaper, F.; Graeve, L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem. J. 1998, 334, 297–314. [Google Scholar] [CrossRef]
- Schindler, C.; Plumlee, C. Inteferons pen the JAK–STAT pathway. Semin. Cell Dev. Biol. 2008, 19, 311–318. [Google Scholar] [CrossRef]
- Caldenhoven, E.; van Dijk, T.B.; Solari, R.; Armstrong, J.; Raaijmakers, J.A.M.; Lammers, J.-W.J.; Koenderman, L.; de Groot, R.P. STAT3beta, a splice variant of transcription factor STAT3, is a dominant negative regulator of transcription. J. Biol. Chem. 1996, 271, 13221–13227. [Google Scholar] [CrossRef]
- Shao, H.; Quintero, A.J.; Tweardy, D.J. Identification and characterization of cis elements in the STAT3 gene regulating STAT3alpha and STAT3beta messenger RNA splicing. Blood 2001, 98, 3853–3856. [Google Scholar] [CrossRef]
- Yoo, J.-Y.; Huso, D.L.; Nathans, D.; Desiderio, S. Specific ablation of Stat3beta distorts the pattern of Stat3-responsive gene expression and impairs recovery from endotoxic shock. Cell 2002, 108, 331–344. [Google Scholar] [CrossRef] [PubMed]
- Maritano, D.; Sugrue, M.L.; Tininini, S.; Dewilde, S.; Strobl, B.; Fu, X.; Murray-Tait, V.; Chiarle, R.; Poli, V. The STAT3 isoforms alpha and beta have unique and specific functions. Nat. Immunol. 2004, 5, 401–409. [Google Scholar] [CrossRef] [PubMed]
- Hoey, T.; Zhang, S.; Schmidt, N.; Yu, Q.; Ramchandani, S.; Xu, X.; Naeger, L.K.; Sun, Y.; Kaplan, M.H. Distinct requirements for the naturally occurring splice forms Stat4alpha and Stat4beta in IL-12 responses. EMBO J. 2003, 22, 4237–4248. [Google Scholar] [CrossRef] [PubMed]
- Morris, R.; Kershaw, N.J.; Babon, J.J. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018, 27, 1984–2009. [Google Scholar] [CrossRef]
- Mowen, K.; David, M. Role of the STAT1-SH2 domain and STAT2 in the activation and nuclear translocation of STAT1. J. Biol. Chem. 1998, 273, 30073–30076. [Google Scholar] [CrossRef]
- Rawlings, J.S.; Rosler, K.M.; Harrison, D.A. The JAK/STAT signaling pathway. J. Cell Sci. 2004, 117, 1281–1283. [Google Scholar] [CrossRef]
- Wen, Z.; Zhong, Z.; Darnell, J.E., Jr. Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 1995, 82, 241–250. [Google Scholar] [CrossRef]
- Goh, K.C.; Haque, S.; Williams, B. P38 MAP kinase is required for STAT1 serine phosphorylation and transcriptional activation induced by interferons. EMBO J. 1999, 18, 5601–5608. [Google Scholar] [CrossRef]
- Kovarik, P.; Stoiber, D.; Novy, M.; Decker, T. Stat1 combines signals derived from IFN-gamma and LPS receptors during macrophage activation. EMBO J. 1998, 17, 3660–3668. [Google Scholar] [CrossRef]
- Jain, N.; Zhang, T.; Fong, S.L.; Lim, C.P.; Cao, X. Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK). Oncogene 1998, 17, 3157–3167. [Google Scholar] [CrossRef]
- Lim, C.P.; Cao, X. Serine phosphorylation and negative regulation of Stat3 by JNK. J. Biol. Chem. 1999, 274, 31055–31061. [Google Scholar] [CrossRef] [PubMed]
- Sengupta, T.K.; Talbot, E.S.; Scherle, P.A.; Ivashkiv, L.B. Rapid inhibition of interleukin-6 signaling and Stat3 activation mediated by mitogen-activated protein kinases. Proc. Natl. Acad. Sci. USA 1998, 95, 11107–11112. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef] [PubMed]
- Gilmour, K.C.; Reich, N.C. Signal transduction and activation of gene transcription by interferons. Gene Expr. 1995, 5, 1–18. [Google Scholar] [PubMed]
- Tassiulas, I.; Hu, X.; Ho, H.; Kashyap, Y.; Paik, P.; Hu, Y.; Lowell, C.A.; Ivashkiv, L.B. Amplification of IFN-alpha-induced STAT1 activation and inflammatory function by Syk and ITAM-containing adaptors. Nat. Immunol. 2004, 5, 1181–1189. [Google Scholar] [CrossRef]
- Cheng, C.; Lin, H.; Tsai, K.; Chiang, Y.; Lim, K.; Chen, C.G.; Su, Y.; Peng, C.; Ho, A.; Huang, L.; et al. Epidermal growth factor induces STAT1 expression to exacerbate the IFNr-mediated PD-L1 axis in epidermal growth factor receptor-positive cancers. Mol. Carcinog. 2018, 57, 1588–1598. [Google Scholar] [CrossRef]
- Costa-Pereira, A.P.; Tininini, S.; Strobl, B.; Alonzi, T.; Schlaak, J.F.; Is’harc, H.; Gesualdo, I.; Newman, S.J.; Kerr, I.M.; Poli, V. Mutational switch of an IL-6 response to an interferon-gamma-like response. Proc. Natl. Acad. Sci. USA 2002, 99, 8043–8047. [Google Scholar] [CrossRef]
- Kalliolias, G.D.; Ivashkiv, L.B. IL-27 activates human monocytes via STAT1 and suppresses IL-10 production but the inflammatory functions of IL-27 are abrogated by TLRs and p38. J. Immunol. 2008, 180, 6325–6333. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, Z. STAT1 in cancer: Friend or foe? Discov. Med. 2017, 24, 19–29. [Google Scholar]
- Leung, S.; Qureshi, S.A.; Kerr, I.M.; Darnell, J.E.; Stark, G.R. Role of STAT2 in the alpha interferon signaling pathway. Mol. Cell Biol. 1995, 15, 1312–1317. [Google Scholar] [CrossRef]
- Lu, W.; Chen, H.; Yel, F.; Wang, F.; Xie, X. VEGF induces phosphorylation of STAT3 through binding VEGFR2 in ovarian carcinoma cells in vitro. Eur. J. Gynaecol. Oncol. 2006, 27, 363–369. [Google Scholar]
- Morinobu, A.; Gadina, M.; Strober, W.; Visconti, R.; Fornace, A.; Montagna, C.; Feldman, G.M.; Nishikomori, R.; O’Shea, J.J. STAT4 serine phosphorylation is critical for IL-12-induced IFN-gamma production but not for cell proliferation. Proc. Natl. Acad. Sci. USA 2002, 99, 12281–12286. [Google Scholar] [CrossRef] [PubMed]
- Alkharusi, A.; AlMuslahi, A.; AlBalushi, N.; AlAjmi, R.; AlRawahi, S.; AlFarqani, A.; Norstedt, G.; Zadjali, F. Connections between prolactin and ovarian cancer. PLoS ONE 2021, 16, e0255701. [Google Scholar] [CrossRef] [PubMed]
- Tsuji-Takayama, K.; Otani, T.; Inoue, T.; Nakamura, S.; Motoda, R.; Kibata, M.; Orita, K. Erythropoietin induces sustained phosphorylation of STAT5 in primitive but not definitive erythrocytes generated from mouse embryonic stem cells. Exp. Hematol. 2006, 34, 1323–1332. [Google Scholar] [CrossRef]
- Waqas, S.F.H.; Ampem, G.; Röszer, T. Analysis of IL-4/STAT6 Signaling in Macrophages. Methods Mol. Biol. 2019, 1966, 211–224. [Google Scholar] [CrossRef] [PubMed]
- Cao, H.; Zhang, J.; Liu, H.; Wan, L.; Zhang, H.; Huang, Q.; Xu, E.; Lai, M. IL-13/STAT6 signaling plays a critical role in the epithelial-mesenchymal transition of colorectal cancer cells. Oncotarget 2016, 7, 61183–61198. [Google Scholar] [CrossRef]
- Xiao, L.; Naganawa, T.; Obugunde, E.; Gronowicz, G.; Ornitz, D.; Coffin, J.D.; Hurley, M.M. Stat1 controls postnatal bone formation by regulating fibroblast growth factor signaling in osteoblasts. J. Biol. Chem. 2004, 279, 27743–27752. [Google Scholar] [CrossRef]
- Shou, P.; Chen, Q.; Jiang, J.; Xu, C.; Zhang, J.; Zheng, C.; Jiang, M.; Velletri, T.; Cao, W.; Huang, Y.; et al. Type I interferons exert anti-tumor effect via reversing immunosuppression mediated by mesenchymal stromal cells. Oncogene 2016, 35, 5953–5962. [Google Scholar] [CrossRef]
- Josahkian, J.A.; Saggioro, F.P.; Vidotto, T.; Ventura, H.T.; dos Reis, F.J.C.; de Sousa, C.B.; Tiezzi, D.G.; de Andrade, J.M.; Koti, M.; Squire, J.A. Increased STAT1 Expression in High Grade Serous Ovarian Cancer Is Associated with a Better Outcome. Int. J. Gynecol. Cancer 2018, 28, 459–465. [Google Scholar] [CrossRef]
- Burke, A.J.; Garrido, P.; Johnson, C.; Sullivan, F.J.; Glynn, S.A.; Johnson, C.; Sullivan, F. Inflammation and Nitrosative Stress Effects in Ovarian and Prostate Pathology and Carcinogenesis. Antioxid. Redox Signal. 2017, 26, 1078–1090. [Google Scholar] [CrossRef]
- Trinh, B.; Ko, S.Y.; Haria, D.; Barengo, N.; Naora, H. The homeoprotein DLX4 controls inducible nitric oxide synthase-mediated angiogenesis in ovarian cancer. Mol. Cancer 2015, 14, 97. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Zhu, L.; Hao, B.; Gao, W.; Wang, Q.; Li, K.; Wang, M.; Huang, M.; Liu, Z.; Yang, Q.; et al. iNOS-derived nitric oxide promotes glycolysis by inducing pyruvate kinase M2 nuclear translocation in ovarian cancer. Oncotarget 2017, 8, 33047–33063. [Google Scholar] [CrossRef] [PubMed]
- Leung, E.L.; Fraser, M.; Fiscus, R.R.; Tsang, B.K. Cisplatin alters nitric oxide synthase levels in human ovarian cancer cells: Involvement in p53 regulation and cisplatin resistance. Br. J. Cancer 2008, 98, 1803–1809. [Google Scholar] [CrossRef] [PubMed]
- Tian, X.; Guan, W.; Zhang, L.; Sun, W.; Zhou, D.; Lin, Q.; Ren, W.; Nadeem, L.; Xu, G. Physical interaction of STAT1 isoforms with TGF-beta receptors leads to functional crosstalk between two signaling pathways in epithelial ovarian cancer. J. Exp. Clin. Cancer Res. 2018, 37, 103. [Google Scholar] [CrossRef] [PubMed]
- Au, K.K.; Le Page, C.; Ren, R.; Meunier, L.; Clément, I.; Tyrishkin, K.; Peterson, N.; Kendall-Dupont, J.; Childs, T.; Francis, J.; et al. STAT1-associated intratumoural T H 1 immunity predicts chemotherapy resistance in high-grade serous ovarian cancer. J. Pathol. Clin. Res. 2016, 2, 259–270. [Google Scholar] [CrossRef]
- Au, K.K.; Peterson, N.; Truesdell, P.; Reid-Schachter, G.; Khalaj, K.; Ren, R.; Francis, J.-A.; Graham, C.H.; Craig, A.W.; Koti, M. CXCL10 alters the tumour immune microenvironment and disease progression in a syngeneic murine model of high-grade serous ovarian cancer. Gynecol. Oncol. 2017, 145, 436–445. [Google Scholar] [CrossRef]
- Duncan, C.J.A.; Hambleton, S. Human Disease Phenotypes Associated with Loss and Gain of Function Mutations in STAT2: Viral Susceptibility and Type I Interferonopathy. J. Clin. Immunol. 2021, 41, 1446–1456. [Google Scholar] [CrossRef]
- Zhong, Z.; Wen, Z.; Darnell, J.E., Jr. Stat3: A STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 1994, 264, 95–98. [Google Scholar] [CrossRef]
- Zhao, S.-H.; Zhao, F.; Zheng, J.-Y.; Gao, L.-F.; Zhao, X.-J.; Cui, M.-H. Knockdown of stat3 expression by RNAi inhibits in vitro growth of human ovarian cancer. Radiol. Oncol. 2011, 45, 196–203. [Google Scholar] [CrossRef]
- Manz, M.; Boettcher, S. Emergency granulopoiesis. Nat. Rev. Immunol. 2014, 14, 302–314. [Google Scholar] [CrossRef]
- Yang, P.-L.; Liu, L.-X.; Li, E.-M.; Xu, L.-Y. STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy. Cancers 2020, 12, 2459. [Google Scholar] [CrossRef] [PubMed]
- Jin, W. Role of JAK/STAT3 Signaling in the Regulation of Metastasis, the Transition of Cancer Stem Cells, and Chemoresistance of Cancer by Epithelial-Mesenchymal Transition. Cells 2020, 9, 217. [Google Scholar] [CrossRef] [PubMed]
- Shih, P.-C.; Mei, K.-C. Role of STAT3 signaling transduction pathways in cancer stem cell-associated chemoresistance. Drug Discov. Today 2021, 26, 1450–1458. [Google Scholar] [CrossRef]
- Wang, X.; Zhang, X.; Qiu, C.; Yang, N. STAT3 Contributes to Radioresistance in Cancer. Front. Oncol. 2020, 10, 1120. [Google Scholar] [CrossRef] [PubMed]
- Selvendiran, K.; Bratasz, A.; Kuppusamy, M.L.; Tazi, M.F.; Rivera, B.K.; Kuppusamy, P. Hypoxia induces chemoresistance in ovarian cancer cells by activation of signal transducer and activator of transcription 3. Int. J. Cancer 2009, 125, 2198–2204. [Google Scholar] [CrossRef]
- Jin, P.; Liu, Y.; Wang, R. STAT3 regulated miR-216a promotes ovarian cancer proliferation and cisplatin resistance. Biosci. Rep. 2018, 38, BSR20180547. [Google Scholar] [CrossRef]
- Li, Q.; Yang, F.; Shi, X.; Bian, S.; Shen, F.; Wu, Y.; Zhu, C.; Fu, F.; Wang, J.; Zhou, J.; et al. MTHFD2 promotes ovarian cancer growth and metastasis via activation of the STAT3 signaling pathway. FEBS Open Bio 2021, 11, 2845–2857. [Google Scholar] [CrossRef]
- Wang, X.; Fu, Y.; Xing, Y. TRIM47 promotes ovarian cancer cell proliferation, migration, and invasion by activating STAT3 signaling. Clinics 2022, 77, 100122. [Google Scholar] [CrossRef]
- Chen, H.; Ye, D.; Xie, X.; Chen, B.; Lu, W. VEGF, VEGFRs expressions and activated STATs in ovarian epithelial carcinoma. Gynecol. Oncol. 2004, 94, 630–635. [Google Scholar] [CrossRef]
- Anglesio, M.S.; George, J.; Kulbe, H.; Friedlander, M.; Rischin, D.; Lemech, C.; Power, J.; Coward, J.; Cowin, P.A.; House, C.M.; et al. IL6-STAT3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Clin. Cancer Res. 2011, 17, 2538–2548. [Google Scholar] [CrossRef]
- Wei, L.-H.; Kuo, M.-L.; Chen, C.-A.; Chou, C.-H.; Lai, K.-B.; Lee, C.-N.; Hsieh, C.-Y. Interleukin-6 promotes cervical tumor growth by VEGF-dependent angiogenesis via a STAT3 pathway. Oncogene 2003, 22, 1517–1527. [Google Scholar] [CrossRef] [PubMed]
- Yin, S.; Yang, L.; Zheng, Y.; Zang, R. Wip1 suppresses angiogenesis through the STAT3-VEGF signalling pathway in serous ovarian cancer. J. Ovarian Res. 2022, 15, 56. [Google Scholar] [CrossRef]
- Martincuks, A.; Song, J.; Kohut, A.; Zhang, C.; Li, Y.-J.; Zhao, Q.; Mak, E.; Rodriguez-Rodriguez, L.; Yu, H.; Cristea, M. PARP Inhibition Activates STAT3 in Both Tumor and Immune Cells Underlying Therapy Resistance and Immunosuppression In Ovarian Cancer. Front. Oncol. 2021, 11, 5163. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef]
- Yue, P.; Zhang, X.; Paladino, D.; Sengupta, B.; Ahmad, S.; Holloway, R.W.; Ingersoll, S.B.; Turkson, J. Hyperactive EGF receptor, Jaks and Stat3 signaling promote enhanced colony-forming ability, motility and migration of cisplatin-resistant ovarian cancer cells. Oncogene 2012, 31, 2309–2322. [Google Scholar] [CrossRef] [PubMed]
- Chong, K.Y.; Kang, M.; Garofalo, F.; Ueno, D.; Liang, H.; Cady, S.; Madarikan, O.; Pitruzzello, N.; Tsai, C.-H.; Hartwich, T.M.; et al. Inhibition of Heat Shock Protein 90 suppresses TWIST1 Transcription. Mol. Pharmacol. 2019, 96, 168–179. [Google Scholar] [CrossRef] [PubMed]
- Zheng, B.; Geng, L.; Zeng, L.; Liu, F.; Huang, Q. AKT2 contributes to increase ovarian cancer cell migration and invasion through the AKT2-PKM2-STAT3/NF-kappaB axis. Cell Signal. 2018, 45, 122–131. [Google Scholar] [CrossRef] [PubMed]
- Rich, J.N. Cancer stem cells: Understanding tumor hierarchy and heterogeneity. Medicine 2016, 95, S2–S7. [Google Scholar] [CrossRef]
- Chen, M.-W.; Yang, S.-T.; Chien, M.-H.; Hua, K.-T.; Wu, C.-J.; Hsiao, S.M.; Lin, H.; Hsiao, M.; Su, J.-L.; Wei, L.-H. The STAT3-miRNA-92-Wnt Signaling Pathway Regulates Spheroid Formation and Malignant Progression in Ovarian Cancer. Cancer Res. 2017, 77, 1955–1967. [Google Scholar] [CrossRef]
- Burgos-Ojeda, D.; Wu, R.; McLean, K.; Chen, Y.-C.; Talpaz, M.; Yoon, E.; Cho, K.R.; Buckanovich, R.J. CD24+ Ovarian Cancer Cells Are Enriched for Cancer-Initiating Cells and Dependent on JAK2 Signaling for Growth and Metastasis. Mol. Cancer Ther. 2015, 14, 1717–1727. [Google Scholar] [CrossRef]
- Ning, Y.; Cui, Y.; Li, X.; Cao, X.; Chen, A.; Xu, C.; Cao, J.; Luo, X. Co-culture of ovarian cancer stem-like cells with macrophages induced SKOV3 cells stemness via IL-8/STAT3 signaling. Biomed. Pharmacother. 2018, 103, 262–271. [Google Scholar] [CrossRef] [PubMed]
- Giordano, M.; Decio, A.; Battistini, C.; Baronio, M.; Bianchi, F.; Villa, A.; Bertalot, G.; Freddi, S.; Lupia, M.; Jodice, M.G.; et al. L1CAM promotes ovarian cancer stemness and tumor initiation via FGFR1/SRC/STAT3 signaling. J. Exp. Clin. Cancer Res. 2021, 40, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.; Mai, H.; Peng, J.; Zhou, B.; Hou, J.; Jiang, D. STAT4: An immunoregulator contributing to diverse human diseases. Int. J. Biol. Sci. 2020, 16, 1575–1585. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Ji, G.; Le, X.; Luo, Z.; Wang, C.; Feng, M.; Xu, L.; Zhang, Y.; Lau, W.B.; Lau, B.; et al. An integrated analysis identifies STAT4 as a key regulator of ovarian cancer metastasis. Oncogene 2017, 36, 3384–3396. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Wang, J.; Chen, W.; Chen, X.; Wang, J. Overexpression of STAT4 under hypoxia promotes EMT through miR-200a/STAT4 signal pathway. Life Sci. 2021, 273, 119263. [Google Scholar] [CrossRef]
- Azam, M.; Erdjument-Bromage, H.; Kreider, B.; Xia, M.; Quelle, F.; Basu, R.; Saris, C.; Tempst, P.; Ihle, J.; Schindler, C. Interleukin-3 signals through multiple isoforms of Stat5. EMBO J. 1995, 14, 1402–1411. [Google Scholar] [CrossRef]
- Schindler, C.; Levy, D.E.; Decker, T. JAK-STAT signaling: From interferons to cytokines. J. Biol. Chem. 2007, 282, 20059–20063. [Google Scholar] [CrossRef]
- Lin, J.-X.; Leonard, W.J. The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene 2000, 19, 2566–2576. [Google Scholar] [CrossRef]
- Warsch, W.; Kollmann, K.; Eckelhart, E.; Fajmann, S.; Cerny-Reiterer, S.; Hölbl, A.; Gleixner, K.V.; Dworzak, M.; Mayerhofer, M.; Hoermann, G.; et al. High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia. Blood 2011, 117, 3409–3420. [Google Scholar] [CrossRef]
- Wu, C.-J.; Sundararajan, V.; Sheu, B.-C.; Huang, R.Y.-J.; Wei, L.-H. Activation of STAT3 and STAT5 Signaling in Epithelial Ovarian Cancer Progression: Mechanism and Therapeutic Opportunity. Cancers 2019, 12, 24. [Google Scholar] [CrossRef]
- Jinawath, N.; Vasoontara, C.; Jinawath, A.; Fang, X.; Zhao, K.; Yap, K.L.; Guo, T.; Lee, C.S.; Wang, W.; Balgley, B.M.; et al. Oncoproteomic analysis reveals co-upregulation of RELA and STAT5 in carboplatin resistant ovarian carcinoma. PLoS ONE 2010, 5, e11198. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Dong, Z.; Li, Y.; Yang, Y.; Yuan, Z.; Qu, X.; Kong, B. The upregulation of signal transducer and activator of transcription 5-dependent microRNA-182 and microRNA-96 promotes ovarian cancer cell proliferation by targeting forkhead box O3 upon leptin stimulation. Int. J. Biochem. Cell Biol. 2013, 45, 536–545. [Google Scholar] [CrossRef] [PubMed]
- Chen, K.-H.E.; Walker, A.M. Prolactin inhibits a major tumor-suppressive function of wild type BRCA1. Cancer Lett. 2016, 375, 293–302. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Kim, J.W.; Choi, D.K.; Yu, J.H.; Kim, J.H.; Lee, D.-S.; Min, S.-H. Poziotinib suppresses ovarian cancer stem cell growth via inhibition of HER4-mediated STAT5 pathway. Biochem. Biophys. Res. Commun. 2020, 526, 158–164. [Google Scholar] [CrossRef]
- Duetsch, G.; Illig, T.; Loesgen, S.; Rohde, K.; Klopp, N.; Herbon, N.; Gohlke, H.; Altmueller, J.; Wjst, M. STAT6 as an asthma candidate gene: Polymorphism-screening, association and haplotype analysis in a Caucasian sib-pair study. Hum. Mol. Genet. 2002, 11, 613–621. [Google Scholar] [CrossRef]
- Gong, M.; Zhuo, X.; Ma, A. STAT6 Upregulation Promotes M2 Macrophage Polarization to Suppress Atherosclerosis. Med. Sci. Monit. Basic Res. 2017, 23, 240–249. [Google Scholar] [CrossRef]
- Takeda, K.; Tanaka, T.; Shi, W.; Matsumoto, M.; Minami, M.; Kashiwamura, S.-I.; Nakanishi, K.; Yoshida, N.; Kishimoto, T.; Akira, S. Essential role of Stat6 in IL-4 signalling. Nature 1996, 380, 627–630. [Google Scholar] [CrossRef]
- Ruan, Z.; Yang, X.; Cheng, W. OCT4 accelerates tumorigenesis through activating JAK/STAT signaling in ovarian cancer side population cells. Cancer Manag. Res. 2019, 11, 389–399. [Google Scholar] [CrossRef]
- Zhou, Z.; Wang, P.; Sun, R.; Li, J.; Hu, Z.; Xin, H.; Luo, C.; Zhou, J.; Fan, J.; Zhou, S. Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J. Immunother. Cancer 2021, 9, e001946. [Google Scholar] [CrossRef]
- Vankerckhoven, A.; Wouters, R.; Mathivet, T.; Ceusters, J.; Baert, T.; Van Hoylandt, A.; Gerhardt, H.; Vergote, I.; Coosemans, A. Opposite Macrophage Polarization in Different Subsets of Ovarian Cancer: Observation from a Pilot Study. Cells 2020, 9, 305. [Google Scholar] [CrossRef]
- Liu, R.; Hu, R.; Zeng, Y.; Zhang, W.; Zhou, H.-H. Tumour immune cell infiltration and survival after platinum-based chemotherapy in high-grade serous ovarian cancer subtypes: A gene expression-based computational study. Ebiomedicine 2020, 51, 102602. [Google Scholar] [CrossRef] [PubMed]
- Ciucci, A.; Zannoni, G.F.; Buttarelli, M.; Martinelli, E.; Mascilini, F.; Petrillo, M.; Ferrandina, G.; Scambia, G.; Gallo, D. Ovarian low and high grade serous carcinomas: Hidden divergent features in the tumor microenvironment. Oncotarget 2016, 7, 68033–68043. [Google Scholar] [CrossRef] [PubMed]
- Takaishi, K.; Komohara, Y.; Tashiro, H.; Ohtake, H.; Nakagawa, T.; Katabuchi, H.; Takeya, M. Involvement of M2-polarized macrophages in the ascites from advanced epithelial ovarian carcinoma in tumor progression via Stat3 activation. Cancer Sci. 2010, 101, 2128–2136. [Google Scholar] [CrossRef] [PubMed]
- Kujawski, M.; Kortylewski, M.; Lee, H.; Herrmann, A.; Kay, H.; Yu, H. Stat3 mediates myeloid cell–dependent tumor angiogenesis in mice. J. Clin. Investig. 2008, 118, 3367–3377. [Google Scholar] [CrossRef]
- Zajac, E.; Schweighofer, B.; Kupriyanova, T.A.; Juncker-Jensen, A.; Minder, P.; Quigley, J.P.; Deryugina, E.I. Angiogenic capacity of M1- and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9. Blood 2013, 122, 4054–4067. [Google Scholar] [CrossRef]
- Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
- Chitu, V.; Stanley, E.R. Colony-stimulating factor-1 in immunity and inflammation. Curr. Opin. Immunol. 2006, 18, 39–48. [Google Scholar] [CrossRef]
- Genin, M.; Clement, F.; Fattaccioli, A.; Raes, M.; Michiels, C. M1 and M2 macrophages derived from THP-1 cells differentially modulate the response of cancer cells to etoposide. BMC Cancer 2015, 15, 577. [Google Scholar] [CrossRef]
- Wang, Q.; Ni, H.; Lan, L.; Wei, X.; Xiang, R.; Wang, Y. Fra-1 protooncogene regulates IL-6 expression in macrophages and promotes the generation of M2d macrophages. Cell Res. 2010, 20, 701–712. [Google Scholar] [CrossRef]
- Reinartz, S.; Schumann, T.; Finkernagel, F.; Wortmann, A.; Jansen, J.M.; Meissner, W.; Krause, M.; Schwörer, A.; Wagner, U.; Müller-Brüsselbach, S.; et al. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: Correlation of CD163 expression, cytokine levels and early relapse. Int. J. Cancer 2014, 134, 32–42. [Google Scholar] [CrossRef]
- Porcheray, F.; Viaud, S.; Rimaniol, A.-C.; Léone, C.; Samah, B.; Dereuddre-Bosquet, N.; Dormont, D.; Gras, G. Macrophage activation switching: An asset for the resolution of inflammation. Clin. Exp. Immunol. 2005, 142, 481–489. [Google Scholar] [CrossRef] [PubMed]
- Cai, D.L.; Jin, L.-P. Immune Cell Population in Ovarian Tumor Microenvironment. J. Cancer 2017, 8, 2915–2923. [Google Scholar] [CrossRef] [PubMed]
- Domínguez-Soto, A.; Sierra-Filardi, E.; Puig-Kröger, A.; Pérez-Maceda, B.; Gómez-Aguado, F.; Corcuera, M.T.; Sánchez-Mateos, P.; Corbí, A.L. Dendritic cell-specific ICAM-3–grabbing nonintegrin expression on M2-polarized and tumor-associated macrophages is macrophage-CSF dependent and enhanced by tumor-derived IL-6 and IL-10. J. Immunol. 2011, 186, 2192–2200. [Google Scholar] [CrossRef] [PubMed]
- Kortylewski, M.; Kujawski, M.; Wang, T.; Wei, S.; Zhang, S.; Pilon-Thomas, S.; Niu, G.; Kay, H.; Mulé, J.; Kerr, W.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat. Med. 2005, 11, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
- Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef]
- Bharadwaj, U.; Li, M.; Zhang, R.; Chen, C.; Yao, Q. Elevated Interleukin-6 and G-CSF in human pancreatic cancer cell conditioned medium suppress dendritic cell differentiation and activation. Cancer Res. 2007, 67, 5479–5488. [Google Scholar] [CrossRef]
- Park, S.-J.; Nakagawa, T.; Kitamura, H.; Atsumi, T.; Kamon, H.; Sawa, S.-I.; Kamimura, D.; Ueda, N.; Iwakura, Y.; Ishihara, K.; et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J. Immunol. 2004, 173, 3844–3854. [Google Scholar] [CrossRef]
- Nefedova, Y.; Huang, M.; Kusmartsev, S.; Bhattacharya, R.; Cheng, P.; Salup, R.; Jove, R.; Gabrilovich, D. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J. Immunol. 2004, 172, 464–474. [Google Scholar] [CrossRef]
- Yu, S.; Liu, C.; Su, K.; Wang, J.; Liu, Y.; Zhang, L.; Li, C.; Cong, Y.; Kimberly, R.; Grizzle, W.E.; et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J. Immunol. 2007, 178, 6867–6875. [Google Scholar] [CrossRef]
- Herrmann, A.; Kortylewski, M.; Kujawski, M.; Zhang, C.; Reckamp, K.; Armstrong, B.; Wang, L.; Kowolik, C.; Deng, J.; Figlin, R.; et al. Targeting Stat3 in the myeloid compartment drastically improves the in vivo antitumor functions of adoptively transferred T cells. Cancer Res. 2010, 70, 7455–7464. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Velders, M.P.; Sotomayor, E.M.; Kast, W.M. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J. Immunol. 2001, 166, 5398–5406. [Google Scholar] [CrossRef] [PubMed]
- Gabrilovich, D.I.; Bronte, V.; Chen, S.-H.; Colombo, M.P.; Ochoa, A.; Ostrand-Rosenberg, S.; Schreiber, H. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007, 67, 425. [Google Scholar] [CrossRef] [PubMed]
- Nagaraj, S.; Nelson, A.; Youn, J.-I.; Cheng, P.; Quiceno, D.; Gabrilovich, D.I. Antigen-specific CD4(+) T cells regulate function of myeloid-derived suppressor cells in cancer via retrograde MHC class II signaling. Cancer Res. 2012, 72, 928–938. [Google Scholar] [CrossRef]
- Diaz-Montero, C.M.; Salem, M.L.; Nishimura, M.I.; Garrett-Mayer, E.; Cole, D.J.; Montero, A.J. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy. Cancer Immunol. Immunother. 2009, 58, 49–59. [Google Scholar] [CrossRef]
- Cheng, P.; Corzo, C.A.; Luetteke, N.; Yu, B.; Nagaraj, S.; Bui, M.M.; Ortiz, M.; Nacken, W.; Sorg, C.; Vogl, T.; et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J. Exp. Med. 2008, 205, 2235–2249. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, T.; Kuniyasu, Y.; Toda, M.; Sakaguchi, N.; Itoh, M.; Iwata, M.; Shimizu, J. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: Induction of autoimmune disease by breaking their anergic/suppressive state. Int. Immunol. 1998, 10, 1969–1980. [Google Scholar] [CrossRef]
- Yu, P.; Lee, Y.; Liu, W.; Krausz, T.; Chong, A.; Schreiber, H.; Fu, Y.-X. Intratumor depletion of CD4+ cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors. J. Exp. Med. 2005, 201, 779–791. [Google Scholar] [CrossRef]
- Martinez-Lostao, L.; Briones, J.; Forné, I.; Gallo, M.M.; Ferrer, B.; Sierra, J.; Rodriguez-Sanchez, J.L.; Juarez, C. Role of the STAT1 pathway in apoptosis induced by fludarabine and JAK kinase inhibitors in B-cell chronic lymphocytic leukemia. Leuk. Lymphoma 2005, 46, 435–442. [Google Scholar] [CrossRef] [PubMed]
- Lukenbill, J.; Kalaycio, M. Fludarabine: A review of the clear benefits and potential harms. Leuk. Res. 2013, 37, 986–994. [Google Scholar] [CrossRef]
- Wallen, H.; Thompson, J.A.; Reilly, J.Z.; Rodmyre, R.M.; Cao, J.; Yee, C. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PLoS ONE 2009, 4, e4749. [Google Scholar] [CrossRef]
- Zhang, L.; Zou, W. Inhibition of integrin beta1 decreases the malignancy of ovarian cancer cells and potentiates anticancer therapy via the FAK/STAT1 signaling pathway. Mol. Med. Rep. 2015, 12, 7869–7876. [Google Scholar] [CrossRef] [PubMed]
- Xue, J.; Bi, X.; Wu, G.; Meng, D.; Fang, J. Fludarabine reduces survivability of HepG2 cells through VEGF under hypoxia. Arch. Biochem. Biophys. 2007, 468, 100–106. [Google Scholar] [CrossRef] [PubMed]
- Zaffaroni, N.; Orlandi, L.; Gornati, D.; De Marco, C.; Vaglini, M.; Silvestrini, R. Fludarabine as a modulator of cisplatin activity in human tumour primary cultures and established cell lines. Eur. J. Cancer 1996, 32, 1766–1773. [Google Scholar] [CrossRef] [PubMed]
- Hoogstad-van Evert, J.; Bekkers, R.; Ottevanger, N.; Schaap, N.; Hobo, W.; Jansen, J.H.; Massuger, L.; Dolstra, H. Intraperitoneal infusion of ex vivo-cultured allogeneic NK cells in recurrent ovarian carcinoma patients (a phase I study). Medicine 2019, 98, e14290. [Google Scholar] [CrossRef]
- Gunning, P.T.; Katt, W.; Glenn, M.; Siddique, K.; Kim, J.S.; Jove, R.; Sebti, S.M.; Turkson, J.; Hamilton, A.D. Isoform selective inhibition of STAT1 or STAT3 homo-dimerization via peptidomimetic probes: Structural recognition of STAT SH2 domains. Bioorganic Med. Chem. Lett. 2007, 17, 1875–1878. [Google Scholar] [CrossRef]
- Zhou, X.-X.; Gao, P.-J.; Sun, B.-G. Pravastatin attenuates interferon-gamma action via modulation of STAT1 to prevent aortic atherosclerosis in apolipoprotein E-knockout mice. Clin. Exp. Pharmacol. Physiol. 2009, 36, 373–379. [Google Scholar] [CrossRef]
- Shuai, K.; Stark, G.R.; Kerr, L.M.; Darnell, J.E. A single phosphotyrosine residue of Stat91 required for gene activation by interferon-gamma. Science 1993, 261, 1744–1746. [Google Scholar] [CrossRef]
- Yeh, C. Pravastatin inhibits tumor growth trhough elevating the levels of apolipoprotein A1. Adv. Dig. Med. 2015, 3, 3–10. [Google Scholar] [CrossRef]
- Irvin, S.; Clarke, M.A.; Trabert, B.; Wentzensen, N. Systematic review and meta-analysis of studies assessing the relationship between statin use and risk of ovarian cancer. Cancer Causes Control 2020, 31, 869–879. [Google Scholar] [CrossRef]
- Chou, P.-H.; Luo, C.-K.; Wali, N.; Lin, W.-Y.; Ng, S.-K.; Wang, C.-H.; Zhao, M.; Lin, S.-W.; Yang, P.-M.; Liu, P.-J.; et al. A chemical probe inhibitor targeting STAT1 restricts cancer stem cell traits and angiogenesis in colorectal cancer. J. Biomed. Sci. 2022, 29, 20. [Google Scholar] [CrossRef]
- Luo, C.-K.; Chou, P.-H.; Ng, S.-K.; Lin, W.-Y.; Wei, T.-T. Cannabinoids orchestrate cross-talk between cancer cells and endothelial cells in colorectal cancer. Cancer Gene Ther. 2022, 29, 597–611. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Wang, J.; Wu, J.; Zheng, Q.; Hu, J. Indirubin suppresses ovarian cancer cell viabilities through the STAT3 signaling pathway. Drug Des. Dev. Ther. 2018, 12, 3335–3342. [Google Scholar] [CrossRef] [PubMed]
- Eisenbrand, G.; Hippe, F.; Jakobs, S.; Muehlbeyer, S. Molecular mechanisms of indirubin and its derivatives: Novel anticancer molecules with their origin in traditional Chinese phytomedicine. J. Cancer Res. Clin. Oncol. 2004, 130, 627–635. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Song, Y.; Wu, Y.; Dong, Y.; Lai, L.; Zhang, J.; Lu, B.; Dai, F.; He, L.; Liu, M.; et al. Indirubin inhibits tumor growth by antitumor angiogenesis via blocking VEGFR2-mediated JAK/STAT3 signaling in endothelial cell. Int. J. Cancer 2011, 129, 2502–2511. [Google Scholar] [CrossRef]
- Kotha, A.; Sekharam, M.; Cilenti, L.; Siddiquee, K.; Khaled, A.; Zervos, A.S.; Carter, B.; Turkson, J.; Jove, R. Resveratrol inhibits Src and Stat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol. Cancer Ther. 2006, 5, 621–629. [Google Scholar] [CrossRef]
- Kohandel, Z.; Farkhondeh, T.; Aschner, M.; Pourbagher-Shahri, A.M.; Samarghandian, S. STAT3 pathway as a molecular target for resveratrol in breast cancer treatment. Cancer Cell Int. 2021, 21, 468. [Google Scholar] [CrossRef]
- Baek, S.H.; Ko, J.-H.; Lee, H.; Jung, J.; Kong, M.; Lee, J.-W.; Lee, J.; Chinnathambi, A.; Zayed, M.; Alharbi, S.A.; et al. Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction and radiosensitization in head and neck tumor cells. Phytomedicine 2016, 23, 566–577. [Google Scholar] [CrossRef]
- Sun, X.; Xu, Q.; Zeng, L.; Xie, L.; Zhao, Q.; Xu, H.; Wang, X.; Jiang, N.; Fu, P.; Sang, M. Resveratrol suppresses the growth and metastatic potential of cervical cancer by inhibiting STAT3 Tyr705 phosphorylation. Cancer Med. 2020, 9, 8685–8700. [Google Scholar] [CrossRef]
- Li, Y.; Zhu, W.; Li, J.; Liu, M.; Wei, M. Resveratrol suppresses the STAT3 signaling pathway and inhibits proliferation of high glucose-exposed HepG2 cells partly through SIRT1. Oncol. Rep. 2013, 30, 2820–2828. [Google Scholar] [CrossRef]
- Zhong, L.-X.; Li, H.; Wu, M.-L.; Liu, X.-Y.; Zhong, M.-J.; Chen, X.-Y.; Liu, J.; Zhang, Y. Inhibition of STAT3 signaling as critical molecular event in resveratrol-suppressed ovarian cancer cells. J. Ovarian Res. 2015, 8, 25. [Google Scholar] [CrossRef]
- Cheuk, I.W.; Chen, J.; Siu, M.; Ho, J.C.; Lam, S.S.; Shin, V.Y.; Kwong, A. Resveratrol enhanced chemosensitivity by reversing macrophage polarization in breast cancer. Clin. Transl. Oncol. 2022, 24, 854–863. [Google Scholar] [CrossRef] [PubMed]
- Alexandrow, M.G.; Song, L.J.; Altiok, S.; Gray, J.; Haura, E.B.; Kumar, N.B. Curcumin: A novel Stat3 pathway inhibitor for chemoprevention of lung cancer. Eur. J. Cancer Prev. 2012, 21, 407–412. [Google Scholar] [CrossRef]
- Glienke, W.; Maute, L.; Wicht, J.; Bergmann, L. Curcumin inhibits constitutive STAT3 phosphorylation in human pancreatic cancer cell lines and downregulation of survivin/BIRC5 gene expression. Cancer Investig. 2010, 28, 166–171. [Google Scholar] [CrossRef] [PubMed]
- Bill, M.A.; Nicholas, C.; Mace, T.A.; Etter, J.P.; Li, C.; Schwartz, E.B.; Fuchs, J.R.; Young, G.S.; Lin, L.; Lin, J.; et al. Structurally modified curcumin analogs inhibit STAT3 phosphorylation and promote apoptosis of human renal cell carcinoma and melanoma cell lines. PLoS ONE 2012, 7, e40724. [Google Scholar] [CrossRef] [PubMed]
- Gong, X.; Jiang, L.; Li, W.; Liang, Q.; Li, Z. Curcumin induces apoptosis and autophagy inhuman renal cell carcinoma cells via Akt/mTOR suppression. Bioengineered 2021, 12, 5017–5027. [Google Scholar] [CrossRef]
- Chung, S.S.; Vadgama, J.V. Curcumin and epigallocatechin gallate inhibit the cancer stem cell phenotype via down-regulation of STAT3-NFkappaB signaling. Anticancer Res. 2015, 35, 39–46. [Google Scholar]
- Seo, J.H.; Jeong, K.J.; Oh, W.J.; Sul, H.J.; Sohn, J.S.; Kim, Y.K.; Cho, D.Y.; Kang, J.K.; Park, C.G.; Lee, H.Y. Lysophosphatidic acid induces STAT3 phosphorylation and ovarian cancer cell motility: Their inhibition by curcumin. Cancer Lett. 2010, 288, 50–56. [Google Scholar] [CrossRef]
- Saydmohammed, M.; Joseph, D.; Syed, V. Curcumin suppresses constitutive activation of STAT-3 by up-regulating protein inhibitor of activated STAT-3 (PIAS-3) in ovarian and endometrial cancer cells. J. Cell. Biochem. 2010, 110, 447–456. [Google Scholar] [CrossRef]
- Kim, M.J.; Park, K.-S.; Kim, K.-T.; Gil, E.Y. The inhibitory effect of curcumin via fascin suppression through JAK/STAT3 pathway on metastasis and recurrence of ovary cancer cells. BMC Women’s Health 2020, 20, 256. [Google Scholar] [CrossRef]
- Ham, I.-H.; Wang, L.; Lee, D.; Woo, J.; Kim, T.H.; Jeong, H.Y.; Oh, H.J.; Choi, K.S.; Hur, H. Curcumin inhibits the cancer-associated fibroblast-derived chemoresistance of gastric cancer through the suppression of the JAK/STAT3 signaling pathway. Int. J. Oncol. 2022, 61, 85. [Google Scholar] [CrossRef]
- Sandhiutami, N.M.D.; Arozal, W.; Louisa, M.; Rahmat, D.; Wuyung, P.E. Curcumin Nanoparticle Enhances the Anticancer Effect of Cisplatin by Inhibiting PI3K/AKT and JAK/STAT3 Pathway in Rat Ovarian Carcinoma Induced by DMBA. Front. Pharmacol. 2021, 11, 603235. [Google Scholar] [CrossRef] [PubMed]
- Dei Cas, M.; Ghidoni, R. Dietary Curcumin: Correlation between Bioavailability and Health Potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef]
- Tabanelli, R.; Brogi, S.; Calderone, V. Improving Curcumin Bioavailability: Current Strategies and Future Perspectives. Pharmaceutics 2021, 13, 1715. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Hutzen, B.; Zuo, M.; Ball, S.; Deangelis, S.; Foust, E.; Pandit, B.; Ihnat, M.A.; Shenoy, S.S.; Kulp, S.; et al. Novel STAT3 phosphorylation inhibitors exhibit potent growth-suppressive activity in pancreatic and breast cancer cells. Cancer Res. 2010, 70, 2445–2454. [Google Scholar] [CrossRef]
- Fossey, S.L.; Bear, M.D.; Lin, J.; Li, C.; Schwartz, E.B.; Li, P.-K.; Fuchs, J.R.; Fenger, J.; Kisseberth, W.C.; London, C.A. The novel curcumin analog FLLL32 decreases STAT3 DNA binding activity and expression, and induces apoptosis in osteosarcoma cell lines. BMC Cancer 2011, 11, 112. [Google Scholar] [CrossRef] [PubMed]
- ElNaggar, A.C.; Saini, U.; Naidu, S.; Wanner, R.; Sudhakar, M.; Fowler, J.; Nagane, M.; Kuppusamy, P.; Cohn, D.E.; Selvendiran, K. Anticancer potential of diarylidenyl piperidone derivatives, HO-4200 and H-4318, in cisplatin resistant primary ovarian cancer. Cancer Biol. Ther. 2016, 17, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
- Rath, K.S.; Naidu, S.K.; Lata, P.; Bid, H.K.; Rivera, B.K.; McCann, G.A.; Tierney, B.J.; ElNaggar, A.C.; Bravo, V.; Leone, G.; et al. HO-3867, a safe STAT3 inhibitor, is selectively cytotoxic to ovarian cancer. Cancer Res. 2014, 74, 2316–2327. [Google Scholar] [CrossRef]
- Bixel, K.; Saini, U.; Bid, H.K.; Fowler, J.; Riley, M.; Wanner, R.; Dorayappan, K.D.P.; Rajendran, S.; Konishi, I.; Matsumura, N.; et al. Targeting STAT3 by HO3867 induces apoptosis in ovarian clear cell carcinoma. Int. J. Cancer 2017, 141, 1856–1866. [Google Scholar] [CrossRef]
- Saini, U.; Naidu, S.; ElNaggar, A.C.; Bid, H.K.; Wallbillich, J.J.; Bixel, K.; Bolyard, C.; A Suarez, A.; Kaur, B.; Kuppusamy, P.; et al. Elevated STAT3 expression in ovarian cancer ascites promotes invasion and metastasis: A potential therapeutic target. Oncogene 2017, 36, 168–181. [Google Scholar] [CrossRef]
- Qian, X.-P.; Zhang, X.-H.; Sun, L.-N.; Xing, W.-F.; Wang, Y.; Sun, S.-Y.; Ma, M.-Y.; Cheng, Z.-P.; Wu, Z.-D.; Xing, C.; et al. Corosolic acid and its structural analogs: A systematic review of their biological activities and underlying mechanism of action. Phytomedicine 2021, 91, 153696. [Google Scholar] [CrossRef]
- Fujiwara, Y.; Takaishi, K.; Nakao, J.; Ikeda, T.; Katabuchi, H.; Takeya, M.; Komohara, Y. Corosolic acid enhances the antitumor effects of chemotherapy on epithelial ovarian cancer by inhibiting signal transducer and activator of transcription 3 signaling. Oncol. Lett. 2013, 6, 1619–1623. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Widjaya, A.S.; Liu, J.; Liu, X.; Long, Z.; Jiang, Y. Cell-penetrating corosolic acid liposome as a functional carrier for delivering chemotherapeutic drugs. Acta Biomater. 2020, 106, 301–313. [Google Scholar] [CrossRef] [PubMed]
- Blaskovich, M.A.; Sun, J.; Cantor, A.; Turkson, J.; Jove, R.; Sebti, S.M. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003, 63, 1270–1279. [Google Scholar]
- Liu, T.; Peng, H.; Zhang, M.; Deng, Y.; Wu, Z. Cucurbitacin B, a small molecule inhibitor of the Stat3 signaling pathway, enhances the chemosensitivity of laryngeal squamous cell carcinoma cells to cisplatin. Eur. J. Pharmacol. 2010, 641, 15–22. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Lu, B.; Zhang, X.; Zhang, J.; Lai, L.; Li, D.; Wu, Y.; Song, Y.; Luo, J.; Pang, X.; et al. Cucurbitacin E, a tetracyclic triterpenes compound from Chinese medicine, inhibits tumor angiogenesis through VEGFR2-mediated Jak2-STAT3 signaling pathway. Carcinogenesis 2010, 31, 2097–2104. [Google Scholar] [CrossRef]
- Shan, X.-L.; Zhou, X.-Y.; Yang, J.; Wang, Y.-L.; Deng, Y.-H.; Zhang, M.-X. Inhibitory effect of cucurbitacin E on the proliferation of ovarian cancer cells and its mechanism. Chin. J. Cancer 2010, 29, 20–24. [Google Scholar] [CrossRef]
- Guo, H.; Kuang, S.; Song, Q.-L.; Liu, M.; Sun, X.-X.; Yu, Q. Cucurbitacin I inhibits STAT3, but enhances STAT1 signaling in human cancer cells in vitro through disrupting actin filaments. Acta Pharmacol. Sin. 2018, 39, 425–437. [Google Scholar] [CrossRef]
- Li, R.; Xiao, J.; Tang, S.; Lin, X.; Xu, H.; Han, B.; Yang, M.; Liu, F. Cucurbitacin I induces apoptosis in ovarian cancer cells through oxidative stress and the p190B-Rac1 signaling axis. Mol. Med. Rep. 2020, 22, 2545–2550. [Google Scholar] [CrossRef]
- Yu, B.; Liang, J.; Li, X.; Liu, L.; Yao, J.; Chen, X.; Chen, R. Renieramycin T Inhibits Melanoma B16F10 Cell Metastasis and Invasion via Regulating Nrf2 and STAT3 Signaling Pathways. Molecules 2022, 27, 5337. [Google Scholar] [CrossRef]
- Liu, Q.; Wang, K. The induction of ferroptosis by impairing STAT3/Nrf2/GPx4 signaling enhances the sensitivity of osteosarcoma cells to cisplatin. Cell Biol. Int. 2019, 43, 1245–1256. [Google Scholar] [CrossRef]
- Hua, Y.; Yuan, X.; Shen, Y.-H.; Wang, J.; Azeem, W.; Yang, S.; Gade, A.; Lellahi, S.M.; Øyan, A.M.; Ke, X.; et al. Novel STAT3 Inhibitors Targeting STAT3 Dimerization by Binding to the STAT3 SH2 Domain. Front. Pharmacol. 2022, 13, 836724. [Google Scholar] [CrossRef] [PubMed]
- Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
- McMurray, J.S. A New Small-Molecule Stat3 Inhibitor. Chem. Biol. 2006, 13, 1123–1124. [Google Scholar] [CrossRef]
- Guo, H.; Xiao, Y.; Yuan, Z.; Yang, X.; Chen, J.; Chen, C.; Wang, M.; Xie, L.; Chen, Q.; Tong, Y.; et al. Inhibition of STAT3Y705 phosphorylation by Stattic suppresses proliferation and induces mitochondrial-dependent apoptosis in pancreatic cancer cells. Cell Death Discov. 2022, 8, 116. [Google Scholar] [CrossRef] [PubMed]
- Pan, Y.; Zhou, F.; Zhang, R.; Claret, F.X. Stat3 inhibitor stattic exhibits potent antitumor activity and induces chemo- and radio-sensitivity in nasopharyngeal carcinoma. PLoS ONE 2013, 8, e54565. [Google Scholar] [CrossRef] [PubMed]
- Méndez-Clemente, A.; Bravo-Cuellar, A.; González-Ochoa, S.; Santiago-Mercado, M.; Palafox-Mariscal, L.; Jave-Suárez, L.; Solorzano-Ibarra, F.; Villaseñor-García, M.; Ortiz-Lazareno, P.; Hernández-Flores, G. Dual STAT-3 and IL-6R inhibition with stattic and tocilizumab decreases migration, invasion and proliferation of prostate cancer cells by targeting the IL-6/IL-6R/STAT-3 axis. Oncol. Rep. 2022, 48, 138. [Google Scholar] [CrossRef] [PubMed]
- Leidgens, V.; Proske, J.; Rauer, L.; Moeckel, S.; Renner, K.; Bogdahn, U.; Riemenschneider, M.J.; Proescholdt, M.; Vollmann-Zwerenz, A.; Hau, P.; et al. Stattic and metformin inhibit brain tumor initiating cells by reducing STAT3-phosphorylation. Oncotarget 2017, 8, 8250–8263. [Google Scholar] [CrossRef]
- Poria, D.K.; Sheshadri, N.; Balamurugan, K.; Sharan, S.; Sterneck, E. The STAT3 inhibitor Stattic acts independently of STAT3 to decrease histone acetylation and modulate gene expression. J. Biol. Chem. 2021, 296, 100220. [Google Scholar] [CrossRef]
- Xia, Y.; Wang, G.; Jiang, M.; Liu, X.; Zhao, Y.; Song, Y.; Jiang, B.; Zhu, D.; Hu, L.; Zhang, Z.; et al. A Novel Biological Activity of the STAT3 Inhibitor Stattic in Inhibiting Glutathione Reductase and Suppressing the Tumorigenicity of Human Cervical Cancer Cells via a ROS-Dependent Pathway. OncoTargets Ther. 2021, 14, 4047–4060. [Google Scholar] [CrossRef]
- 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. USA 2005, 102, 4700–4705. [Google Scholar] [CrossRef]
- Fuh, B.; Sobo, M.; Cen, L.; Josiah, D.; Hutzen, B.; Cisek, K.; Bhasin, D.; Regan, N.; Lin, L.; Chan, C.; et al. LLL-3 inhibits STAT3 activity, suppresses glioblastoma cell growth and prolongs survival in a mouse glioblastoma model. Br. J. Cancer 2009, 100, 106–112. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Hutzen, B.; Li, P.-K.; Ball, S.; Zuo, M.; DeAngelis, S.; Foust, E.; Sobo, M.; Friedman, L.; Bhasin, D.; et al. A novel small molecule, LLL12, inhibits STAT3 phosphorylation and activities and exhibits potent growth-suppressive activity in human cancer cells. Neoplasia 2010, 12, 39–50. [Google Scholar] [CrossRef] [PubMed]
- Mencalha, A.L.; Du Rocher, B.; Salles, D.; Binato, R.; Abdelhay, E. LLL-3, a STAT3 inhibitor, represses BCR-ABL-positive cell proliferation, activates apoptosis and improves the effects of Imatinib mesylate. Cancer Chemother. Pharmacol. 2010, 65, 1039–1046. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Benson, D.M.; DeAngelis, S.; Bakan, C.E.; Li, P.-K.; Li, C.; Lin, J. A small molecule, LLL12 inhibits constitutive STAT3 and IL-6-induced STAT3 signaling and exhibits potent growth suppressive activity in human multiple myeloma cells. Int. J. Cancer 2012, 130, 1459–1469. [Google Scholar] [CrossRef] [PubMed]
- Pan, L.; Chen, X.; Rassool, F.V.; Li, C.; Lin, J. LLL12B, a Novel Small-Molecule STAT3 Inhibitor, Induces Apoptosis and Suppresses Cell Migration and Tumor Growth in Triple-Negative Breast Cancer Cells. Biomedicines 2022, 10, 2003. [Google Scholar] [CrossRef]
- Chen, X.; Pan, L.; Wei, J.; Zhang, R.; Yang, X.; Song, J.; Bai, R.-Y.; Fu, S.; Pierson, C.R.; Finlay, J.L.; et al. LLL12B, a small molecule STAT3 inhibitor, induces growth arrest, apoptosis, and enhances cisplatin-mediated cytotoxicity in medulloblastoma cells. Sci. Rep. 2021, 11, 6517. [Google Scholar] [CrossRef]
- Zhang, R.; Yang, X.; Roque, D.M.; Li, C.; Lin, J. A novel small molecule LLL12B inhibits STAT3 signaling and sensitizes ovarian cancer cell to paclitaxel and cisplatin. PLoS ONE 2021, 16, e0240145. [Google Scholar] [CrossRef]
- Vageli, D.P.; Doukas, P.G.; Siametis, A.; Judson, B.L. Targeting STAT3 prevents bile reflux-induced oncogenic molecular events linked to hypopharyngeal carcinogenesis. J. Cell. Mol. Med. 2022, 26, 75–87. [Google Scholar] [CrossRef]
- Zhang, R.; Chen, X.; Fu, S.; Xu, L.; Lin, J. A small molecule STAT3 inhibitor, LLL12, enhances cisplatin- and paclitaxel-mediated inhibition of cell growth and migration in human ovarian cancer cells. Oncol. Rep. 2020, 44, 1224–1232. [Google Scholar] [CrossRef]
- Leong, P.L.; Andrews, G.A.; Johnson, D.E.; Dyer, K.F.; Xi, S.; Mai, J.C.; Robbins, P.D.; Gadiparthi, S.; Burke, N.A.; Watkins, S.F.; et al. Targeted inhibition of Stat3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc. Natl. Acad. Sci. USA 2003, 100, 4138–4143. [Google Scholar] [CrossRef]
- Xi, S.; E Gooding, W.; Grandis, J.R. In vivo antitumor efficacy of STAT3 blockade using a transcription factor decoy approach: Implications for cancer therapy. Oncogene 2005, 24, 970–979. [Google Scholar] [CrossRef] [PubMed]
- Sen, M.; Tosca, P.J.; Zwayer, C.; Ryan, M.J.; Johnson, J.D.; Knostman, K.A.B.; Giclas, P.C.; Peggins, J.O.; Tomaszewski, J.E.; McMurray, T.P.; et al. Lack of toxicity of a STAT3 decoy oligonucleotide. Cancer Chemother. Pharmacol. 2009, 63, 983–995. [Google Scholar] [CrossRef] [PubMed]
- Sen, M.; Thomas, S.M.; Kim, S.; Yeh, J.I.; Ferris, R.L.; Johnson, J.T.; Duvvuri, U.; Lee, J.; Sahu, N.; Joyce, S.; et al. First-in-human trial of a STAT3 decoy oligonucleotide in head and neck tumors: Implications for cancer therapy. Cancer Discov. 2012, 2, 694–705. [Google Scholar] [CrossRef]
- Vighi, E.; Montanari, M.; Ruozi, B.; Iannuccelli, V.; Leo, E. The role of protamine amount in the transfection performance of cationic SLN designed as a gene nanocarrier. Drug Deliv. 2012, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Severino, P.; Szymanski, M.; Favaro, M.; Azzoni, A.R.; Chaud, M.V.; Santana, M.H.A.; Silva, A.M.; Souto, E.B. Development and characterization of a cationic lipid nanocarrier as non-viral vector for gene therapy. Eur. J. Pharm. Sci. 2015, 66, 78–82. [Google Scholar] [CrossRef] [PubMed]
- A Harrington, H.; Ho, K.L.; Ghosh, S.; Tung, K. Construction and analysis of a modular model of caspase activation in apoptosis. Theor. Biol. Med. Model. 2008, 5, 26. [Google Scholar] [CrossRef]
- Zhang, X.; Lu, T.; Ma, Y.; Li, R.; Pang, Y.; Mao, H.; Liu, P. Novel Nanocomplexes Targeting STAT3 Demonstrate Promising Anti-Ovarian Cancer Effects in vivo. OncoTargets Ther. 2020, 13, 5069–5082. [Google Scholar] [CrossRef]
- Huang, W.; Liu, Y.; Wang, J.; Yuan, X.; Jin, H.-W.; Zhang, L.-R.; Zhang, J.-T.; Liu, Z.-M.; Cui, J.-R. Small-molecule compounds targeting the STAT3 DNA-binding domain suppress survival of cisplatin-resistant human ovarian cancer cells by inducing apoptosis. Eur. J. Med. Chem. 2018, 157, 887–897. [Google Scholar] [CrossRef]
- Bharadwaj, U.; Eckols, T.K.; Xu, X.; Kasembeli, M.M.; Chen, Y.; Adachi, M.; Song, Y.; Mo, Q.; Lai, S.Y.; Tweardy, D.J. Small-molecule inhibition of STAT3 in radioresistant head and neck squamous cell carcinoma. Oncotarget 2016, 7, 26307–26330. [Google Scholar] [CrossRef]
- Kasembeli, M.M.; Singhmar, P.; Ma, J.; Edralin, J.; Tang, Y.; Adams, C.; Heijnen, C.J.; Kavelaars, A.; Tweardy, D.J. TTI-101: A competitive inhibitor of STAT3 that spares oxidative phosphorylation and reverses mechanical allodynia in mouse models of neuropathic pain. Biochem. Pharmacol. 2021, 192, 114688. [Google Scholar] [CrossRef]
- Nelson, E.A.; Walker, S.R.; Weisberg, E.; Bar-Natan, M.; Barrett, R.; Gashin, L.B.; Terrell, S.; Klitgaard, J.L.; Santo, L.; Addorio, M.R.; et al. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood 2011, 117, 3421–3429. [Google Scholar] [CrossRef] [PubMed]
- Nelson, E.A.; Walker, S.R.; Xiang, M.; Weisberg, E.; Bar-Natan, M.; Barrett, R.; Liu, S.; Kharbanda, S.; Christie, A.L.; Nicolais, M.; et al. The STAT5 Inhibitor Pimozide Displays Efficacy in Models of Acute Myelogenous Leukemia Driven by FLT3 Mutations. Genes Cancer 2012, 3, 503–511. [Google Scholar] [CrossRef]
- Zhou, W.; Chen, M.-K.; Yu, H.-T.; Zhong, Z.-H.; Cai, N.; Chen, G.-Z.; Zhang, P.; Chen, J.-J. The antipsychotic drug pimozide inhibits cell growth in prostate cancer through suppression of STAT3 activation. Int. J. Oncol. 2016, 48, 322–328. [Google Scholar] [CrossRef] [PubMed]
- Subramaniam, D.; Angulo, P.; Ponnurangam, S.; Dandawate, P.; Ramamoorthy, P.; Srinivasan, P.; Iwakuma, T.; Weir, S.J.; Chastain, K.; Anant, S. Suppressing STAT5 signaling affects osteosarcoma growth and stemness. Cell Death Dis. 2020, 11, 149. [Google Scholar] [CrossRef] [PubMed]
- Xiao, Z.; Liang, J.; Deng, Q.; Song, C.; Yang, X.; Liu, Z.; Shao, Z.; Zhang, K.; Wang, X.; Li, Z. Pimozide augments bromocriptine lethality in prolactinoma cells and in a xenograft model via the STAT5/cyclin D1 and STAT5/Bcl-xL signaling pathways. Int. J. Mol. Med. 2021, 47, 113–124. [Google Scholar] [CrossRef]
- Natarajan, K.; Müller-Klieser, D.; Rubner, S.; Berg, T. Stafia-1: A STAT5a-Selective Inhibitor Developed via Docking-Based Screening of in Silico O-Phosphorylated Fragments. Chem. A Eur. J. 2020, 26, 148–154. [Google Scholar] [CrossRef]
- Elumalai, N.; Berg, A.; Natarajan, K.; Scharow, A.; Berg, T. Nanomolar Inhibitors of the Transcription Factor STAT5b with High Selectivity over STAT5a. Angew. Chem. Int. Ed. 2015, 54, 4758–4763. [Google Scholar] [CrossRef] [PubMed]
- Elumalai, N.; Berg, A.; Rubner, S.; Blechschmidt, L.; Song, C.; Natarajan, K.; Matysik, J.; Berg, T. Rational development of Stafib-2: A selective, nanomolar inhibitor of the transcription factor STAT5b. Sci. Rep. 2017, 7, 819. [Google Scholar] [CrossRef] [PubMed]
- Wingelhofer, B.; Maurer, B.; Heyes, E.C.; Cumaraswamy, A.A.; Berger-Becvar, A.; De Araujo, E.D.; Orlova, A.; Freund, P.; Ruge, F.; Park, J.; et al. Pharmacologic inhibition of STAT5 in acute myeloid leukemia. Leukemia 2018, 32, 1135–1146. [Google Scholar] [CrossRef]
- Seipel, K.; Graber, C.; Flückiger, L.; Bacher, U.; Pabst, T. Rationale for a Combination Therapy with the STAT5 Inhibitor AC-4-130 and the MCL1 Inhibitor S63845 in the Treatment of FLT3-Mutated or TET2-Mutated Acute Myeloid Leukemia. Int. J. Mol. Sci. 2021, 22, 8092. [Google Scholar] [CrossRef]
- de los Fayos Alonso, I.G.; Zujo, L.; Wiest, I.; Kodajova, P.; Timelthaler, G.; Edtmayer, S.; Zrimšek, M.; Kollmann, S.; Giordano, C.; Kothmayer, M.; et al. PDGFRbeta promotes oncogenic progression via STAT3/STAT5 hyperactivation in anaplastic large cell lymphoma. Mol. Cancer 2022, 21, 172. [Google Scholar] [CrossRef] [PubMed]
- Hadzijusufovic, E.; Keller, A.; Berger, D.; Greiner, G.; Wingelhofer, B.; Witzeneder, N.; Ivanov, D.; Pecnard, E.; Nivarthi, H.; Schur, F.K.M.; et al. STAT5 is Expressed in CD34+/CD38− Stem Cells and Serves as a Potential Molecular Target in Ph-Negative Myeloproliferative Neoplasms. Cancers 2020, 12, 1021. [Google Scholar] [CrossRef]
- Chiba, Y.; Todoroki, M.; Nishida, Y.; Tanabe, M.; Misawa, M. A novel STAT6 inhibitor AS1517499 ameliorates antigen-induced bronchial hypercontractility in mice. Am. J. Respir. Cell Mol. Biol. 2009, 41, 516–524. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.-J.; Kim, K.; Kim, M.; Ahn, Y.-H.; Kang, J.L. Inhibition of STAT6 Activation by AS1517499 Inhibits Expression and Activity of PPARgamma in Macrophages to Resolve Acute Inflammation in Mice. Biomolecules 2022, 12, 447. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Han, Z.; Wang, F.; Qiao, J. The STAT6 inhibitor AS1517499 reduces the risk of asthma in mice with 2,4-dinitrochlorobenzene-induced atopic dermatitis by blocking the STAT6 signaling pathway. Allergy Asthma Clin. Immunol. 2022, 18, 12. [Google Scholar] [CrossRef]
- Liu, L.; Peng, S.; Duan, M.; Liu, C.; Li, L.; Zhang, X.; Ren, B.; Tian, H. The role of C/EBP homologous protein (CHOP) in regulating macrophage polarization in RAW264.7 cells. Microbiol. Immunol. 2021, 65, 531–541. [Google Scholar] [CrossRef]
- Binnemars-Postma, K.; Bansal, R.; Storm, G.; Prakash, J. Targeting the Stat6 pathway in tumor-associated macrophages reduces tumor growth and metastatic niche formation in breast cancer. FASEB J. 2018, 32, 969–978. [Google Scholar] [CrossRef]
- Mendoza-Rodríguez, M.; Sánchez-Barrera, C.Á.; Callejas, B.E.; García-Castillo, V.; Beristain-Terrazas, D.L.; Delgado-Buenrostro, N.L.; Chirino, Y.I.; León-Cabrera, S.A.; Rodríguez-Sosa, M.; Gutierrez-Cirlos, E.B.; et al. Use of STAT6 Phosphorylation Inhibitor and Trimethylglycine as New Adjuvant Therapies for 5-Fluorouracil in Colitis-Associated Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 2130. [Google Scholar] [CrossRef]
- Siemasko, K.; Chong, A.S.-F.; Jack, H.-M.; Gong, H.; Williams, J.W.; Finnegan, A. Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the immunosuppressive drug leflunomide leads to a block in IgG1 production. J. Immunol. 1998, 160, 1581–1588. [Google Scholar] [CrossRef]
- Breedveld, F.C. Leflunomide: Mode of action in the treatment of rheumatoid arthritis. Ann. Rheum. Dis. 2000, 59, 841–849. [Google Scholar] [CrossRef]
- Xu, X.; Sun, J.; Song, R.; Doscas, M.E.; Williamson, A.J.; Zhou, J.; Sun, J.; Jiao, X.; Liu, X.; Li, Y. Inhibition of p70 S6 kinase (S6K1) activity by A77 1726, the active metabolite of leflunomide, induces autophagy through TAK1-mediated AMPK and JNK activation. Oncotarget 2017, 8, 30438–30454. [Google Scholar] [CrossRef] [PubMed]
- Doscas, M.E.; Williamson, A.J.; Usha, L.; Bogachkov, Y.; Rao, G.S.; Xiao, F.; Wang, Y.; Ruby, C.; Kaufman, H.; Zhou, J.; et al. Inhibition of p70 S6 kinase (S6K1) activity by A77 1726 and its effect on cell proliferation and cell cycle progress. Neoplasia 2014, 16, 824–834. [Google Scholar] [CrossRef] [PubMed]
- Di Stefano, A.; Iovino, F.; Lombardo, Y.; Eterno, V.; Höger, T.; Dieli, F.; Stassi, G.; Todaro, M. Survivin is regulated by interleukin-4 in colon cancer stem cells. J. Cell. Physiol. 2010, 225, 555–561. [Google Scholar] [CrossRef] [PubMed]
- Stegeman, H.; Kaanders, J.H.; Verheijen, M.M.; Peeters, W.J.; Wheeler, D.L.; Iida, M.; Grénman, R.; van der Kogel, A.J.; Span, P.N.; Bussink, J. Combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition reduces survival of head and neck cancer lines. Mol. Cancer 2013, 12, 133. [Google Scholar] [CrossRef] [PubMed]
- Dietrich, S.; Krämer, O.H.; Hahn, E.; Schäfer, C.; Giese, T.; Hess, M.; Tretter, T.; Rieger, M.; Hüllein, J.; Zenz, T.; et al. Leflunomide induces apoptosis in fludarabine-resistant and clinically refractory CLL cells. Clin. Cancer Res. 2012, 18, 417–431. [Google Scholar] [CrossRef] [PubMed]
- Buglio, D.; Georgakis, G.V.; Hanabuchi, S.; Arima, K.; Khaskhely, N.M.; Liu, Y.-J.; Younes, A. Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood 2008, 112, 1424–1433. [Google Scholar] [CrossRef] [PubMed]
- Munshi, A.; Tanaka, T.; Hobbs, M.L.; Tucker, S.L.; Richon, V.M.; Meyn, R.E. Vorinostat, a histone deacetylase inhibitor, enhances the response of human tumor cells to ionizing radiation through prolongation of gamma-H2AX foci. Mol. Cancer Ther. 2006, 5, 1967–1974. [Google Scholar] [CrossRef] [PubMed]
- Sarfstein, R.; Bruchim, I.; Fishman, A.; Werner, H. The mechanism of action of the histone deacetylase inhibitor vorinostat involves interaction with the insulin-like growth factor signaling pathway. PLoS ONE 2011, 6, e24468. [Google Scholar] [CrossRef]
- Palmieri, D.; Lockman, P.R.; Thomas, F.C.; Hua, E.; Herring, J.; Hargrave, E.; Johnson, M.; Flores, N.; Qian, Y.; Vega-Valle, E.; et al. Vorinostat inhibits brain metastatic colonization in a model of triple-negative breast cancer and induces DNA double-strand breaks. Clin. Cancer Res. 2009, 15, 6148–6157. [Google Scholar] [CrossRef]
- Sakurai, M.; Nishio, M.; Yamamoto, K.; Okuda, T.; Kawano, K.; Ohnuki, T. TMC-264, a novel inhibitor of STAT6 activation produced by phoma sp. TC 1674. J. Antibiot. 2003, 56, 513–519. [Google Scholar] [CrossRef]
- Lai, D.; Wang, A.; Cao, Y.; Zhou, K.; Mao, Z.; Dong, X.; Tian, J.; Xu, D.; Dai, J.; Peng, Y.; et al. Bioactive Dibenzo-alpha-pyrone Derivatives from the Endophytic Fungus Rhizopycnis vagum Nitaf22. J. Nat. Prod. 2016, 79, 2022–2031. [Google Scholar] [CrossRef]
- Feng, Z.; Zheng, W.; Tang, Q.; Cheng, L.; Li, H.; Ni, W.; Pan, X. Fludarabine inhibits STAT1-mediated up-regulation of caspase-3 expression in dexamethasone-induced osteoblasts apoptosis and slows the progression of steroid-induced avascular necrosis of the femoral head in rats. Apoptosis 2017, 22, 1001–1012. [Google Scholar] [CrossRef] [PubMed]
- Bharti, A.C.; Donato, N.; Aggarwal, B.B. Curcumin (diferuloylmethane) inhibits constitutive and IL-6-inducible STAT3 phosphorylation in human multiple myeloma cells. J. Immunol. 2003, 171, 3863–3871. [Google Scholar] [CrossRef] [PubMed]
- Fujiwara, Y.; Komohara, Y.; Ikeda, T.; Takeya, M. Corosolic acid inhibits glioblastoma cell proliferation by suppressing the activation of signal transducer and activator of transcription-3 and nuclear factor-kappa B in tumor cells and tumor-associated macrophages. Cancer Sci. 2011, 102, 206–211. [Google Scholar] [CrossRef]
- Zhang, L.; Wang, Y.; Dong, Y.; Chen, Z.; Eckols, T.K.; Kasembeli, M.M.; Tweardy, D.J.; Mitch, W.E. Pharmacokinetics and pharmacodynamics of TTI-101, a STAT3 inhibitor that blocks muscle proteolysis in rats with chronic kidney disease. Am. J. Physiol. Physiol. 2020, 319, F84–F92. [Google Scholar] [CrossRef] [PubMed]
- Whitley, M.P. Treatment dilemma: A political science perspective. J. Psychosoc. Nurs. Ment. Health Serv. 1991, 29, 35–45. [Google Scholar] [CrossRef] [PubMed]
Proteins | Activators | Biologic Response | References |
---|---|---|---|
STAT1 | IFNα, IFNβ, IFNγ, IFNλ, EGF, PDGF, IL6, IL27 | Promote inflammation, PD-L1 expression, apoptosis, MYC gene expression, monocyte activation | [45,46,47,48,49] |
STAT2 | IFNα, IFNβ, IFNλ | Promote inflammation | [50] |
STAT3 | IL6, IL6 family cytokines, IL10, IL22, Prolactin, Growth Hormone, EGF, TGFα, VEGF | Promote proliferation, chemoresistance, cell migration | [51] |
STAT4 | IFNγ, IL12, IL23, IL2, IL35 | Promote T Cell differentiation, IFNγ production, NK cell activation | [52] |
STAT5 | Growth hormone, Prolactin, IFNα, IFNβ, IL3, Thrombopoietin, Erythropoietin, GM-CSF | Promote proliferation, cell migration | [53,54] |
STAT6 | IL4, IL13 | Promote macrophage polarization, EMT | [55,56] |
Protein | Inhibitors | Interacting Site | References |
---|---|---|---|
STAT1 | Fludarabine | N/A | [252] |
ISS840 | STAT1 SH2 domain | [145] | |
Pravastatin | N/A | [146] | |
THIF | STAT1 SH2 domain | [150] | |
STAT2 | N/A | N/A | N/A |
STAT3 | Indirubin | C-Src, CDK, GSK3β, STAT3 DB domain | [152,154] |
Resveratrol | Src | [155] | |
Curcumin | STAT3 CC domain | [253] | |
FLL31/32 | JAK2, STAT3 SH2 domain | [175] | |
HO-3867/4200 | STAT3 DB domain | [175] | |
H-4318 | STAT3 DB domain | [176] | |
Corosolic acid | N/A | [254] | |
Cucurbitacin -I, -B, -E | JAK2, STAT3 | [183,184,185] | |
323-1/323-2 | STAT3 SH2 domain | [191] | |
Stattic | STAT3 SH2 domain | [192] | |
STA-21 | STAT3 DB domain | [200] | |
LLL-3/LLL-12 | STAT3 SH2 domain | [201] | |
LC28 | STAT3 SH2 domain | [218] | |
TTI-101 | STAT3 SH2 domain | [220,255] | |
STAT4 | N/A | N/A | N/A |
STAT5 | Pimozide | N/A | [221] |
Stafia-1 | STAT5a SH2 domain | [226] | |
Stafib-1/2 | STAT5b SH2 domain | [227,228] | |
AC-4-130 | STAT5 SH2 domain | [229] | |
STAT6 | AS1517499 | N/A | [256] |
Leflunomide | STAT6 DB domain | [239] | |
Vorinostat | N/A | [246] | |
TMC-264 | STAT6 DB domain | [250] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Standing, D.; Feess, E.; Kodiyalam, S.; Kuehn, M.; Hamel, Z.; Johnson, J.; Thomas, S.M.; Anant, S. The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers 2023, 15, 2485. https://doi.org/10.3390/cancers15092485
Standing D, Feess E, Kodiyalam S, Kuehn M, Hamel Z, Johnson J, Thomas SM, Anant S. The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers. 2023; 15(9):2485. https://doi.org/10.3390/cancers15092485
Chicago/Turabian StyleStanding, David, Emma Feess, Satvik Kodiyalam, Michael Kuehn, Zachary Hamel, Jaimie Johnson, Sufi Mary Thomas, and Shrikant Anant. 2023. "The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy" Cancers 15, no. 9: 2485. https://doi.org/10.3390/cancers15092485
APA StyleStanding, D., Feess, E., Kodiyalam, S., Kuehn, M., Hamel, Z., Johnson, J., Thomas, S. M., & Anant, S. (2023). The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers, 15(9), 2485. https://doi.org/10.3390/cancers15092485