|
參考文獻 1.Kim JE, et al. STAT3 Activation in Glioblastoma: Biochemical and Therapeutic Implications. Cancers (Basel) 2014; 6:376-95. 2.White AC, et al. Refining the role for adult stem cells as cancer cells of origin. Trends cell biol 2015; 25:11-20. 3.Jovčevska I, et al. Glioma and glioblastoma - how much do we (not) know? Mol Clin Oncol 2013; 1:935-41. 4.Linos E, et al. Atopy and risk of brain tumors: a meta-analysis. Journal of the National Cancer Institute 2007; 99:1544-50. 5.Kleihues P, et al. Primary and secondary glioblastomas: from concept to clinical diagnosis. Neuro-oncology 1999; 1:44-51. 6.Hartmann C, et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta neuropathological 2010; 120:707-18. 7.Ohgaki H, et al. Epidemiology and etiology of gliomas. Acta neuropathological 2005; 109:93-108. 8.Thakkar JP, et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomarkers Prev 2014; 23:1985-96. 9.Bondy ML, et al. Brain tumor epidemiology: consensus from the Brain Tumor Epidemiology Consortium. Cancer 2008; 113:1953-68. 10.Wrensch M, et al. Epidemiology of primary brain tumors: current concepts and review of the literature. Neuro-oncology 2002; 4:278-99. 11.Adamson C, et al. Glioblastoma multiforme: a review of where we have been and where we are going. Expert Opin Investig Drugs 2009; 18(8):1061-83.
12.Golla H, et al. lioblastoma multiforme from diagnosis to death: a prospective, hospital-based, cohort, pilot feasibility study of patient reported symptoms and needs. Support care cancer 2014; 22:3341-52. 13.Sizoo EM, et al. Symptoms and problems in the end-of-life phase of high-grade glioma patients. Neuro-oncology 2010; 12:1162-6. 14.Davis ME. Glioblastoma: Overview of Disease and Treatment. Clin J Oncol Nurs 2016; 20:S2-S8. 15.Ostrom QT, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007-2011. Neuro-oncology 2014; 16:iv1-63. 16.Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002; 359(9311):1011-8. 17. Li S, et al. Polymethoxyflavones: Chemistry, biological activity, and occurrence in orange peel. ACS Symposium Series 2008; 987:191-210. 18.Wang X, et al. Anti-inflammatory effects of polymethoxyflavones from citrus peels: A review. J. Food Bioact 2018; 3:76–86. 19.Tung Y.C, et al. Polymethoxyflavones: Chemistry and molecular mechanisms for cancer prevention and treatment. Curr Pharmacol Rep 2019; 5:98–113. 20.Lai CS, et al. Potent anti-cancer effects of citrus peel flavonoids in human prostate xenograft tumors. Food Funct 2013; 4:944-9. 21.Wang L, et al. Anticancer activities of citrus peel polymethoxyflavones related to angiogenesis and others. Biomed Res Int 2014; 2014:453972. 22.Li S, et al. Efficient and scalable method in isolation of polymethoxyflavones from orange peel extract by supercritical fluid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 846:291-7. 23.Chiu SP, et al. Neurotrophic action of 5-hydroxylated polymethoxyflavones: 5-demethylnobiletin and gardenin A stimulate neuritogenesis in PC12 cells. J Agric Food Chem 2013; 61:9453-63. 24.Li S, et al. Isolation and syntheses of polymethoxyflavones and hydroxylatedpolymethoxyflavones as inhibitors of HL-60 cell lines. Bioorg Med Chem 2007; 15:3381-9. 25.Ram M. Uckoo, et al. Rapid separation method of polymethoxyflavones from citrus using flash chromatography. Sep Purif Technol 2011; 81:151-158. 26.Sundaram R, et al. Tangeretin, a polymethoxylated flavone, modulates lipid homeostasis and decreases oxidative stress by inhibiting NF-κB activation and proinflammatory cytokines in cardiac tissue of streptozotocin-induced diabetic rats. J Funct Foods 2015; 16:315-333. 27.Shu Z, et al. Tangeretin exerts anti-neuroinflammatory effects via NF-κB modulation in lipopolysaccharide-stimulated microglial cells. Int Immunopharmacol 2014; 19:275-282. 28.Periyasamy K, et al. Antitumor efficacy of tangeretin by targeting the oxidative stress mediated on 7,12-dimethylbenz(a) anthracene-induced proliferative breast cancer in Sprague-Dawley rats. Cancer Chemother Pharmacol 2015; 75:263-272. 29.Ma N, et al. 5-Demethyltangeretin is more potent than tangeretin in inhibiting dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA)-induced skin tumorigenesis. J Funct Foods 2014; 11:528-537. 30.Wang J, et al. Pro-apoptotic effects of the novel tangeretin derivate 5-acetyl-6,7,8,4′-tetramethylnortangeretin on MCF-7 breast cancer cells. Cell Biochem Biophys 2014; 70:1255-1263. 31.Lai C.S., et al. Inhibitory effects of 5-demethyltangeretin and 5-acetyloxy-6,7,8,4′-tetramethoxyflavone on human colon cancer cells. Nutri Funct Prop Food 2013; 281-290. 32. Li Y.R, et al. Tangeretin derivative, 5-acetyloxy-6,7,8,4′-tetramethoxyflavone induces G2/M arrest, apoptosis and autophagy in human non-small cell lung cancer cells in vitro and in vivo. Cancer Biol Ther 2015; 17:48-64. 33.Zhi D, et al. 5-Acetyl-6,7,8,4′-tetramethylnortangeretin induces apoptosis in multiple myeloma U266 cells. Food Sci Hum Wellness 2014; 3:197-203. 34.Zhi D, et al. 5-Acetyl-6,7,8,4′-tetramethylnortangeretin induces apoptosis in multiplemyeloma U266 cells. Food Sci Hum Wellness 2014; 3:197-203 35.Wang J, et al. Pro-apoptotic effects of the novel tangeretin derivate 5-acetyl-6,7,8,4′-tetramethylnortangeretin on MCF-7 breast cancer cells. Cell Biochem Biophys 2014; 70:1255-63. 36.Darnell JE, et al. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular proteins. Science 1994; 264:1415–1421. 37.Zhuang, S. Regulation of STAT signaling by acetylation Cell. Signalling 2013; 25: 1924-1931. 38.Furtek SL, et al. Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem Biol 2016; 11:308-18. 39.Maritano D, et al. The STAT3 isoforms alpha and beta have unique and specific functions. Nat Immunol 2014; 5:401-409. 40.Claudinon J, et al. Palmitoylation of interferon‐alpha (IFN‐alpha) receptor subunit IFNAR1 is required for the activation of Stat1 and Stat2 by IFN‐alpha. J Biol Chem 2009; 284:24328-24340. 41.Berclaz G, et al. EGFR dependent expression of STAT3 (but not STAT1) in breast cancer. Int J Oncol 2001; 19:1155-1160. 42.Cao X, et al. Activation and association of Stat3 with Src in v–Src‐transformed cell lines. Mol Cell Biol 1996; 16:1595-1603. 43.Hemmann U, et al. Differential activation of acute phase response factor/Stat3 and Stat1 via the cytoplasmic domain of the interleukin 6 signal transducer gp130. II. Src homology SH2 domains define the specificity of stat factor activation. J Biol Chem 1996; 271:12999-13007. 44.Schust, J, et al. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem Biol 2006; 13:1235-1242. 45.Debnath, B, et al. Small molecule inhibitors of signal transducer and activator of transcription 3 (Stat3) protein. J Med Chem 2012; 55:6645-6668. 46.Luwor, R, et al. The role of Stat3 in glioblastoma multiforme. J Clin Neurosci 2013; 20:907- 911 47.Zhang, X, et al. A novel small-molecule disrupts Stat3 SH2 domain-phosphotyrosine interactions and Stat3-dependent tumor processes. Biochem Pharmacol 2010; 79:1398-1409 48.Bromberg J. Stat proteins and oncogenesis. J Clin Invest 2002; 109:1139–1142. 49.Corvinus, F.M., et al., Persistent STAT3 Activation in Colon Cancer Is Associated with Enhanced Cell Proliferation and Tumor Growth. Neoplasia 2005; 7:545-555. 50.Mohamed, A, et al. The Incidence, Correlation with Tumor Infiltrating Inflammation, and Prognosis of p-STAT3 Expression in Human Gliomas. Clin Cancer Res 2008; 14:8228-8235. 51.Xie, TX, et al. Activation of stat3 in human melanoma promotes brain metastasis. Cancer Res 2006; 66:3188-96. 52.Bao JJ, et al. Inhibition of constitutively active STAT3 by WP1066 suppresses proliferation and induces apoptosis in pancreatic cancer cells. Clin Cancer Res 2005; 11:9026S-7S. 53.Chan KS, et al. Disruption of Stat3 reveals a critical role in both the initiation and the promotion stages of epithelial carcinogenesis. J Clin Invest 2004; 114:720-8. 54.Iwamaru A, et al. A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene 2006; 26:2435-44. 55.Leong PL, et al. Targeted inhibition of Stat3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc Natl Acad Sci U S A 2003; 100:4138-43. 56.Tang GS, et al. Effects of STAT3 antisense oligodeoxynucleotides on apoptosis and proliferation of mouse melanoma cell line B16. Ai Zheng 2006; 25:269-74. 57.Li GH, et al. Knockdown of STAT3 expression by RNAi suppresses growth and induces apoptosis and differentiation in glioblastoma stem cells. Int J Oncol 2010; 37:103-10. 58.Carro MS, et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature 2010; 463:318-25. 59.Wang H, et al. Targeting Interleukin 6 Signaling Suppresses Glioma Stem Cell Survival and Tumor Growth. Stem Cells 2009; 27:2393-404. 60.Lin GS, et al. STAT3 Tyr705 phosphorylation affects clinical outcome in patients with newly diagnosed supratentorial glioblastoma. Med Oncol 2014; 31:924. 61.Rodney B.L, et al. The role of Stat3 in glioblastoma multiforme. J Clin Neurosci 2013; 20:907-911. 62.Sushil G R, et al. Janus kinases: components of multiple signaling pathways. Oncogene 2000; 19:5662-5679. 63.Haan C, et al. Jaks and cytokine receptors — an intimate relationship. Biochem Pharmacol 2006; 72:1538-1546. 64.Buchert M, et al. Targeting JAK kinase in solid tumors: emerging opportunities and challenges. Oncogene 2016; 35:939-951. 65.Ernst M, et al. Acquiring signalling specificity from the cytokine receptor gp130. Trends Genet 2004; 20:23-32. 66.Daniel E, et al. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol 2018; 15: 234-248. 67.Colamonici O, et al. Jak-Stat Signaling: From Basics to Disease. Mol Cell 1994; 14:8133-8142. 68.Sakai I, Nabell L and Kraft AS. Signal transduction by a CD16/CD7/Jak2 fusion protein. J Biol Chem 1995; 270: 18420-18427. 69.Nakamura N, Chin H, Miyasaka N and Miura O. An Epidermal Growth Factor Receptor/Jak2 Tyrosine Kinase Domain Chimera Induces Tyrosine Phosphorylation of Stat5 and Transduces a Growth Signal in Hematopoietic Cells. J Biol Chem 1996; 271: 19483-19488. 70.Kralovics R, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005; 352:1779-1790. 71. Jones AV, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood 2005; 106:2162-2168. 72.Walters DK, et al. Activating alleles of JAK3 in acute megakaryoblastic leukemia. Cancer Cell 2006; 10:65-75. 73.Bose P, Verstovsek S. JAK2 inhibitors for myeloproliferative neoplasms: what is next? Blood 2017; 130:115-125. 74.Kerr JF, et al. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26:239-57. 75.Formigli L, et al. Aponecrosis: morphological and biochemical exploration of a syncretic process of cell death sharing apoptosis and necrosis. Cell Physiol 2000; 182:41-9. 76.Sperandio S, et al. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci U S A 2000; 97:14376-81.
77.Debnath J, et al. Does autophagy contribute to cell death. Autophagy 2005; 1:66-74. 78.Norbury CJ, Hickson ID. Cellular responses to DNA damage. Annu Rev Pharmacol Toxicol 2001; 41:367-401. 79.Hirsch T, et al. The apoptosis-necrosis paradox. Apoptogenic proteases activated after mitochondrial permeability transition determine the mode of cell death. Oncogene 1997; 15:1573-81. 80.Zeiss CJ. The apoptosis-necrosis continuum: insights from genetically altered mice. Vet Pathol 2003; 40:481-95. 81.Susan Elmore. Apoptosis: A Review of Programmed Cell Death. Toxicol Pathol. Author manuscript. Toxicol Pathol 2007; 35:495-516. 82.Desagher, S. and J.C. Martinou, Mitochondria as the central control point of apoptosis. Trends Cell Biol 2000; 10:369-77. 83.Elmore, S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35:495-516. 84.Thomas, et al. Apoptosis Triggers Specific, Rapid, and Global mRNA Decay with 3'' Uridylated Intermediates Degraded by DIS3L2. Cell Reports 2015; 11: 1079-89. 85.Böhm I. Disruption of the cytoskeleton after apoptosis induction by autoantibodies. Autoimmunity 2003; 36:183-189. 86. Susin, S, et al. Two Distinct Pathways Leading to Nuclear Apoptosis. J Exp Med 2000; 192: 571-80. 87.Madeleine Kihlmark, et al. Sequential degradation of proteins from the nuclear envelope during apoptosis. J Cell Sci 2001; 114:3643-53. 88.Nagata S. Apoptotic DNA fragmentation. Exp Cell Res 2000; 256:12-8.
89.Gong JP, et al. A selective procedure for DNA extraction from apoptotic cells applicable for gel electrophoresis and flow cytometry. Anal Biochem 1994; 218: 314-319. 90.Muchmore SW, et al. X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. Nature 1996; 381:335-41. 91.Youle, Richard J. The BCL-2 protein family: opposing activities that mediate cell death. Nature Reviews Molecular Cell Biology 2008; 9:47-59. 92.Coultas, L. and A. Strasser, The role of the Bcl-2 protein family in cancer. Semin Cancer Biol 2003; 13:115-23. 93.Reed JC, et al. Structure-function analysis of Bcl-2 family proteins. Regulators of programmed cell death. Med Biol 1996; 406:99-112. 94.Pearson, J.R.D.; Regad, T. Targeting cellular pathways in glioblastoma multiforme. Signal Transduct. Target Ther 2017; 2;17040. 95.Linder, B, et al. Therapeutic targeting of Stat3 using lipopolyplex nanoparticle-formulated siRNA in a syngeneic orthotopic mouse glioma model. Cancers 2019; 11:333. 96.Jensen, K.V, et al. The JAK2/STAT3 inhibitor pacritinib e ectively inhibits patient-derived GBM brain tumor initiating cells in vitro and when used in combination with temozolomide increases survival in an orthotopic xenograft model. PLoS ONE 2017; 12:e0189670. 97.Lee, B, et al. The polymethoxylated flavone, Tangeretin improves cognitive memory in rats experiencing a single episode of prolonged post-traumatic stress. Anim. Cells Syst 2018; 22:54-62. 98.Wu, C, et al. Tangeretin protects human brain microvascularendothelial cells against oxygen-glucose deprivation-induced injury. J Cell Biochem 2019; 120:4883-4891. 99.Braidy, N, et al. Neuroprotective Effects of Citrus Fruit-Derived Flavonoids, Nobiletin and Tangeretin in Alzheimer''s and Parkinson''s Disease. CNS Neurol Disord Drug Targets 2017; 16:387-397. 100.Datla, K.P, et al. Tissue distribution and neuroprotective ects of citrus flavonoid tangeretin in a rat model of Parkinson’s disease. Neuroreport 2001; 12:3871-3875. 101.Okuyama, S, et al. Permeation of polymethoxyflavones into the mouse brain and their ect on MK-801-induced locomotive hyperactivity. Int J Mol Sci 2017; 18:489. 102.Yuan, G, et al. CucurbitacinI induces protective autophagy in glioblastoma in vitro and in vivo. J Biol Chem 2014; 289:10607-10619. 103.Ouédraogo, Z.G, et al. Role of STAT3 in genesis and progression of human malignant gliomas. Mol Neurobiol 2017; 54:5780-5797. 104.Masliantsev, K, et al. Impact of STAT3 phosphorylation in glioblastoma stem cells radiosensitization and patient outcome. Oncotarget 2018; 9:3968-3979. 105.Ganguly, D, et al. The critical role that STAT3 plays in glioma-initiating cells: STAT3 addiction in glioma. Oncotarget 2018; 9:22095-22112. 106.Luwor, R.B, et al. The role of Stat3 in glioblastoma multiforme. J Clin Neurosci 2013; 20:907-911
|