請用此 Handle URI 來引用此文件:
http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/84881
完整後設資料紀錄
DC 欄位 | 值 | 語言 |
---|---|---|
dc.contributor.advisor | 郭靜穎(Ching-Ying Kuo) | |
dc.contributor.author | Yu-Wen Huang | en |
dc.contributor.author | 黃鈺雯 | zh_TW |
dc.date.accessioned | 2023-03-19T22:30:36Z | - |
dc.date.copyright | 2022-10-05 | |
dc.date.issued | 2022 | |
dc.date.submitted | 2022-08-29 | |
dc.identifier.citation | Chapter 8 –References Cohen, S., Epidermal growth factor. In Vitro Cell Dev Biol, 1987. 23(4): p. 239-46. Cohen, S., Origins of growth factors: NGF and EGF. The Journal of biological chemistry, 2008. 283(49): p. 33793-33797. Schlessinger, J., Cell Signaling by Receptor Tyrosine Kinases. Cell, 2000. 103(2): p. 211-225. Appert-Collin, A., et al., Role of ErbB Receptors in Cancer Cell Migration and Invasion. Frontiers in Pharmacology, 2015. 6(283). Thariat, J., L. Milas, and K.K. Ang, Integrating Radiotherapy With Epidermal Growth Factor Receptor Antagonists and Other Molecular Therapeutics for the Treatment of Head and Neck Cancer. International Journal of Radiation Oncology*Biology*Physics, 2007. 69(4): p. 974-984. Burgess, A.W., et al., An Open-and-Shut Case? Recent Insights into the Activation of EGF/ErbB Receptors. Molecular Cell, 2003. 12(3): p. 541-552. Kim, Y.-N., P. Dam, and P.J. Bertics, Caveolin-1 Phosphorylation in Human Squamous and Epidermoid Carcinoma Cells: Dependence on ErbB1 Expression and Src Activation. Experimental Cell Research, 2002. 280(1): p. 134-147. Yamauchi, T., et al., Tyrosine phosphorylation of the EGF receptor by the kinase Jak2 is induced by growth hormone. Nature, 1997. 390(6655): p. 91-6. Hemmings, B.A. and D.F. Restuccia, PI3K-PKB/Akt pathway. Cold Spring Harbor perspectives in biology, 2012. 4(9): p. a011189-a011189. Kumar, R., H.M. Shepard, and J. Mendelsohn, Regulation of phosphorylation of the c-erbB-2/HER2 gene product by a monoclonal antibody and serum growth factor(s) in human mammary carcinoma cells. Mol Cell Biol, 1991. 11(2): p. 979-86. Otília Menyhárt, L.S.a.B.G., A Comprehensive Outline of Trastuzumab Resistance Biomarkers in HER2 Overexpressing Breast Cancer. Current Cancer Drug Targets, 2015. Zandi, R., et al., Mechanisms for oncogenic activation of the epidermal growth factor receptor. Cellular Signalling, 2007. 19(10): p. 2013-2023. Alessi, D.R., et al., Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol, 1997. 7(4): p. 261-9. Vander Haar, E., et al., Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol, 2007. 9(3): p. 316-23. Ruiz-Saenz, A., et al., HER2 Amplification in Tumors Activates PI3K/Akt Signaling Independent of HER3. Cancer Research, 2018. 78(13): p. 3645-3658. Scaltriti, M. and J. Baselga, The epidermal growth factor receptor pathway: a model for targeted therapy. Clin Cancer Res, 2006. 12(18): p. 5268-72. Liu, X., et al., Development of Effective Therapeutics Targeting HER3 for Cancer Treatment. Biological Procedures Online, 2019. 21(1): p. 1480-9222. Das, P.M., et al., Reactivation of epigenetically silenced HER4/ERBB4 results in apoptosis of breast tumor cells. Oncogene, 2010. 29(37): p. 5214-5219. Yan, M., et al., HER2 aberrations in cancer: implications for therapy. Cancer Treat Rev, 2014. 40(6): p. 770-80. Parida, P.K., et al., Metabolic diversity within breast cancer brain-tropic cells determines metastatic fitness. Cell Metabolism, 2022. 34(1): p. 90-105.e7. Genentech USA, I. The impact of HER2+ cancer. 2022; Available from: https://www.herceptin.com/hcp/treating-HER2-cancer.html. Garnock-Jones, K.P., G.M. Keating, and L.J. Scott, Trastuzumab. Drugs, 2010. 70(2): p. 215-239. Gollamudi, J., et al., Neoadjuvant therapy for early-stage breast cancer: the clinical utility of pertuzumab. Cancer Manag Res, 2016. 8: p. 21-31. Hudis, C.A., Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med, 2007. 357(1): p. 39-51. Yuan, Y., et al., NDUFA4L2 promotes trastuzumab resistance in HER2-positive breast cancer. Ther Adv Med Oncol, 2021. 13: p. 1758-8340. Jiang, J., et al., Type IIB DNA topoisomerase is downregulated by trastuzumab and doxorubicin to synergize cardiotoxicity. Oncotarget, 2017. 9: p. 6095-6108. Mohan, N., J. Jiang, and W.J. Wu, Implications of Autophagy and Oxidative Stress in Trastuzumab-Mediated Cardiac Toxicities. Austin pharmacology & pharmaceutics, 2017. 2(1): p. 1005. Bang, Y.-J., et al., Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. The Lancet, 2010. 376(9742): p. 687-697. Cobleigh, M.A., et al., Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol, 1999. 17(9): p. 2639-48. Rugo, H.S., et al., Effect of a Proposed Trastuzumab Biosimilar Compared With Trastuzumab on Overall Response Rate in Patients With ERBB2 (HER2)-Positive Metastatic Breast Cancer: A Randomized Clinical Trial. Jama, 2017. 317(1): p. 37-47. Wolff, A.C., N.M. Tung, and L.A. Carey, Implications of Neoadjuvant Therapy in Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. J Clin Oncol, 2019. 37(25): p. 2189-2192. Baselga, J., Clinical trials of Herceptin(trastuzumab). Eur J Cancer, 2001. 37 Suppl 1: p. S18-24. Nagy, P., et al., Decreased accessibility and lack of activation of ErbB2 in JIMT-1, a herceptin-resistant, MUC4-expressing breast cancer cell line. Cancer Res, 2005. 65(2): p. 473-82. Narayan, M., et al., Trastuzumab-induced HER reprogramming in 'resistant' breast carcinoma cells. Cancer Res, 2009. 69(6): p. 2191-4. Nahta, R., et al., Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res, 2005. 65(23): p. 11118-28. Nahta, R., et al., P27(kip1) down-regulation is associated with trastuzumab resistance in breast cancer cells. Cancer Res, 2004. 64(11): p. 3981-6. Zhao, Y., et al., Overcoming Trastuzumab Resistance in Breast Cancer by Targeting Dysregulated Glucose Metabolism. Cancer Research, 2011. 71(13): p. 4585-4597. Warburg, O.H., The metabolism of tumours: investigations from the Kaiser Wilhelm Institute for Biology, Berlin-Dahlem. 1930: Constable & Company Limited. Vander Heiden, M.G., L.C. Cantley, and C.B. Thompson, Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science (New York, N.Y.), 2009. 324(5930): p. 1029-1033. Owen, O.E., S.C. Kalhan, and R.W. Hanson, The Key Role of Anaplerosis and Cataplerosis for Citric Acid Cycle Function *. Journal of Biological Chemistry, 2002. 277(34): p. 30409-30412. DeBerardinis, R.J., et al., Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci U S A, 2007. 104(49): p. 19345-50. Bak, D.W., et al., Cysteine reactivity across the subcellular universe. Current Opinion in Chemical Biology, 2019. 48: p. 96-105. Liou, G.-Y. and P. Storz, Reactive oxygen species in cancer. Free radical research, 2010. 44(5): p. 479-496. Mosharov, E., M.R. Cranford, and R. Banerjee, The Quantitatively Important Relationship between Homocysteine Metabolism and Glutathione Synthesis by the Transsulfuration Pathway and Its Regulation by Redox Changes. Biochemistry, 2000. 39(42): p. 13005-13011. Shin, C.-S., et al., The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility. Nature Communications, 2017. 8(1): p. 15074. Fournier, M., et al., Implication of the glutamate–cystine antiporter xCT in schizophrenia cases linked to impaired GSH synthesis. npj Schizophrenia, 2017. 3(1): p. 31. Ignoul, S. and J. Eggermont, CBS domains: structure, function, and pathology in human proteins. American Journal of Physiology-Cell Physiology, 2005. 289(6): p. C1369-C1378. Sbodio, J.I., S.H. Snyder, and B.D. Paul, Regulators of the transsulfuration pathway. Br J Pharmacol, 2019. 176(4): p. 583-593. Sen, S., et al., Role of cystathionine β-synthase in human breast Cancer. Free Radical Biology and Medicine, 2015. 86: p. 228-238. Liu, N., X. Lin, and C. Huang, Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. British Journal of Cancer, 2020. 122(2): p. 279-292. Stephanie J Sacharow, M., Jonathan D Picker, MBChB, PhD, and Harvey L Levy, MD, Homocystinuria Caused by Cystathionine BetaSynthase Deficiency. GeneReviews, 2004 Jan 15. Cai, W.J., et al., Hydrogen sulfide induces human colon cancer cell proliferation: role of Akt, ERK and p21. Cell Biol Int, 2010. 34(6): p. 565-72. Yin, P., et al., Sp1 is involved in regulation of cystathionine γ-lyase gene expression and biological function by PI3K/Akt pathway in human hepatocellular carcinoma cell lines. Cell Signal, 2012. 24(6): p. 1229-40. Wang, L., et al., Cystathionine‑γ‑lyase promotes the metastasis of breast cancer via the VEGF signaling pathway. Int J Oncol, 2019. 55(2): p. 473-487. Guo, W., et al., Cystathionine γ-lyase deficiency aggravates obesity-related insulin resistance via FoxO1-dependent hepatic gluconeogenesis. Faseb j, 2019. 33(3): p. 4212-4224. Zhong, H. and H. Yin, Role of lipid peroxidation derived 4-hydroxynonenal (4-HNE) in cancer: focusing on mitochondria. Redox Biol, 2015. 4: p. 193-9. Koundouros, N. and G. Poulogiannis, Reprogramming of fatty acid metabolism in cancer. British Journal of Cancer, 2020. 122(1): p. 4-22. Yuan, Z.-h., et al., Fatty Acids Metabolism: The Bridge Between Ferroptosis and Ionizing Radiation. Frontiers in Cell and Developmental Biology, 2021. 9(1691). Friedmann Angeli, J.P., et al., Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nature Cell Biology, 2014. 16(12): p. 1180-1191. Yang, Wan S., et al., Regulation of Ferroptotic Cancer Cell Death by GPX4. Cell, 2014. 156(1): p. 317-331. Kawabata, H., Transferrin and transferrin receptors update. Free Radic Biol Med, 2019. 133: p. 46-54. Andrews, N.C. and P.J. Schmidt, Iron homeostasis. Annu Rev Physiol, 2007. 69: p. 69-85. Wenzel, S.E., et al., PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals. Cell, 2017. 171(3): p. 628-641.e26. Ingold, I., et al., Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell, 2018. 172(3): p. 409-422.e21. Shah, R., M.S. Shchepinov, and D.A. Pratt, Resolving the Role of Lipoxygenases in the Initiation and Execution of Ferroptosis. ACS Central Science, 2018. 4(3): p. 387-396. Sato, M., et al., The ferroptosis inducer erastin irreversibly inhibits system xc− and synergizes with cisplatin to increase cisplatin’s cytotoxicity in cancer cells. Scientific Reports, 2018. 8(1): p. 968. Xie, Y., et al., Ferroptosis: process and function. Cell Death & Differentiation, 2016. 23(3): p. 369-379. Stockwell, B.R., et al., Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell, 2017. 171(2): p. 273-285. Gaschler, M.M., et al., FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nature Chemical Biology, 2018. 14(5): p. 507-515. Yang, W.S. and B.R. Stockwell, Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol, 2016. 26(3): p. 165-176. Piro, G., et al., An FGFR3 Autocrine Loop Sustains Acquired Resistance to Trastuzumab in Gastric Cancer Patients. Clin Cancer Res, 2016. 22(24): p. 6164-6175. Mootha, V.K., et al., PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genetics, 2003. 34(3): p. 267-273. Subramanian, A., et al., Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences, 2005. 102(43): p. 15545-15550. Kute, T., et al., Development of Herceptin resistance in breast cancer cells. Cytometry A, 2004. 57(2): p. 86-93. Yoo, H.-C. and J.-M. Han, Amino Acid Metabolism in Cancer Drug Resistance. Cells, 2022. 11(1): p. 140. Li, J., et al., Ferroptosis: past, present and future. Cell Death & Disease, 2020. 11(2): p. 88. Fu, C., et al., Rehmannioside A improves cognitive impairment and alleviates ferroptosis via activating PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway after ischemia. Journal of Ethnopharmacology, 2022. 289: p. 115021. Claret, F. and T. Vu, Trastuzumab: Updated Mechanisms of Action and Resistance in Breast Cancer. Frontiers in Oncology, 2012. 2. Zimmer, A.S., A.E.D. Van Swearingen, and C.K. Anders, HER2-positive breast cancer brain metastasis: A new and exciting landscape. Cancer Rep (Hoboken), 2022. 5(4): p. e1274. Curigliano, G., et al., Tucatinib versus placebo added to trastuzumab and capecitabine for patients with pretreated HER2+ metastatic breast cancer with and without brain metastases (HER2CLIMB): final overall survival analysis. Ann Oncol, 2022. 33(3): p. 321-329. Herceptin (trastuzumab) Label - Accessdata.fda.gov. 1998: p. 33. Wong, H., et al., Integrating Molecular Mechanisms and Clinical Evidence in the Management of Trastuzumab Resistant or Refractory HER‐2+ Metastatic Breast Cancer. The Oncologist, 2011. 16(11): p. 1535-1546. Arribas, J., et al., p95HER2 and Breast Cancer. Cancer Research, 2011. 71(5): p. 1515-1519. Shiau, J.P., et al., Impacts of Oxidative Stress and PI3K/AKT/mTOR on Metabolism and the Future Direction of Investigating Fucoidan-Modulated Metabolism. Antioxidants (Basel), 2022. 11(5). A Phase I Study of MK-2206 in Combination With Trastuzumab and Lapatinib in HER2-Positive Breast and Gastric Cancer. October 12, 2012, National Cancer Institute (NCI). Hudis, C., et al., A phase 1 study evaluating the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors. Breast Cancer Res, 2013. 15(6): p. R110. Khalil, H.S., et al., NRF2 Regulates HER2 and HER3 Signaling Pathway to Modulate Sensitivity to Targeted Immunotherapies. Oxidative Medicine and Cellular Longevity, 2016. 2016: p. 4148791. DeNicola, G.M., et al., Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature, 2011. 475(7354): p. 106-109. Sporn, M.B. and K.T. Liby, NRF2 and cancer: the good, the bad and the importance of context. Nature Reviews Cancer, 2012. 12(8): p. 564-571. Sun, L., et al., Herceptin induces ferroptosis and mitochondrial dysfunction in H9c2 cells. Int J Mol Med, 2022. 49(2). Wang, S., et al., A novel circular RNA confers trastuzumab resistance in human epidermal growth factor receptor 2-positive breast cancer through regulating ferroptosis. Environ Toxicol, 2022. 37(7): p. 1597-1607. Peng, G., et al., Glutathione peroxidase 4 maintains a stemness phenotype, oxidative homeostasis and regulates biological processes in Panc‑1 cancer stem‑like cells. Oncol Rep, 2019. 41(2): p. 1264-1274. Hangauer, M.J., et al., Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature, 2017. 551(7679): p. 247-250. Habib, E., et al., Expression of xCT and activity of system xc− are regulated by NRF2 in human breast cancer cells in response to oxidative stress. Redox Biology, 2015. 5: p. 33-42. Ge, C., et al., The down-regulation of SLC7A11 enhances ROS induced P-gp over-expression and drug resistance in MCF-7 breast cancer cells. Scientific Reports, 2017. 7(1): p. 3791. Lin, W., et al., SLC7A11/xCT in cancer: biological functions and therapeutic implications. Am J Cancer Res, 2020. 10(10): p. 3106-3126. Liu, P., et al., NRF2 regulates the sensitivity of human NSCLC cells to cystine deprivation-induced ferroptosis via FOCAD-FAK signaling pathway. Redox Biology, 2020. 37: p. 101702. Cho, K., et al., Therapeutic Nanoparticles for Drug Delivery in Cancer. Clinical Cancer Research, 2008. 14(5): p. 1310-1316. Kawasaki, E.S. and A. Player, Nanotechnology, nanomedicine, and the development of new, effective therapies for cancer. Nanomedicine, 2005. 1(2): p. 101-9. Prokop, A. and J.M. Davidson, Nanovehicular intracellular delivery systems. Journal of pharmaceutical sciences, 2008. 97(9): p. 3518-3590. Danquah, M.K., X.A. Zhang, and R.I. Mahato, Extravasation of polymeric nanomedicines across tumor vasculature. Adv Drug Deliv Rev, 2011. 63(8): p. 623-39. Maheshwari, N., et al., Chapter 3 - Guiding Factors and Surface Modification Strategies for Biomaterials in Pharmaceutical Product Development, in Biomaterials and Bionanotechnology, R.K. Tekade, Editor. 2019, Academic Press. p. 57-87. 李寶玉、顏寧、魏予全, 奈米生醫材料. 2006, 五南出版社. 202-218. Mitchell, M.J., et al., Engineering precision nanoparticles for drug delivery. Nature Reviews Drug Discovery, 2021. 20(2): p. 101-124. Chenthamara, D., et al., Therapeutic efficacy of nanoparticles and routes of administration. Biomaterials Research, 2019. 23(1): p. 20. Bangham, A.D., Surrogate cells or trojan horses. The discovery of liposomes. BioEssays, 1995. 17(12): p. 1081-1088. Gabizon, A., H. Shmeeda, and Y. Barenholz, Pharmacokinetics of pegylated liposomal Doxorubicin: review of animal and human studies. Clin Pharmacokinet, 2003. 42(5): p. 419-36. Harris, J.M., N.E. Martin, and M. Modi, Pegylation: a novel process for modifying pharmacokinetics. Clin Pharmacokinet, 2001. 40(7): p. 539-51. McLachlan, A.A. and D.G. Marangoni, Interactions between zwitterionic and conventional anionic and cationic surfactants. Journal of Colloid and Interface Science, 2006. 295(1): p. 243-248. Nishiyama, N. and K. Kataoka, Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery. Pharmacol Ther, 2006. 112(3): p. 630-48. Abbasi, E., et al., Dendrimers: synthesis, applications, and properties. Nanoscale Research Letters, 2014. 9(1): p. 247. Klajnert, B. and M. Bryszewska, Dendrimers: properties and applications. Acta Biochim Pol, 2001. 48(1): p. 199-208. Slavin, Y.N., et al., Metal nanoparticles: understanding the mechanisms behind antibacterial activity. Journal of Nanobiotechnology, 2017. 15(1): p. 65. Wang, A.Z., R. Langer, and O.C. Farokhzad, Nanoparticle delivery of cancer drugs. Annu Rev Med, 2012. 63: p. 185-98. Cortes, J. and C. Saura, Nanoparticle albumin-bound (nab™)-paclitaxel: improving efficacy and tolerability by targeted drug delivery in metastatic breast cancer. European Journal of Cancer Supplements, 2010. 8(1): p. 1-10. Fu, Q., et al., Nanoparticle albumin-bound (NAB) technology is a promising method for anti-cancer drug delivery. Recent Pat Anticancer Drug Discov, 2009. 4(3): p. 262-72. Wilczewska, A.Z., et al., Nanoparticles as drug delivery systems. Pharmacological Reports, 2012. 64(5): p. 1020-1037. Pudlarz, A. and J. Szemraj, Nanoparticles as carriers of proteins, peptides and other therapeutic molecules. Open Life Sciences, 2018. 13(1): p. 285-298. Akbarzadeh, A., et al., Liposome: classification, preparation, and applications. Nanoscale Res Lett, 2013. 8(1): p. 102. Damberg, P., J. Jarvet, and A. Gräslund, Micellar Systems as Solvents in Peptide and Protein Structure Determination, in Nuclear Magnetic Resonance of Biological Macromolecules - Part B. 2001. p. 271-285. Reis, C.P., et al., Drug carriers for oral delivery of peptides and proteins: accomplishments and future perspectives. Therapeutic Delivery, 2013. 4(2), 251–265. Allen, T.M. and P.R. Cullis, Drug Delivery Systems: Entering the Mainstream. Science, 2004. 303(5665): p. 1818-1822. Chung, J.E., et al., Self-assembled micellar nanocomplexes comprising green tea catechin derivatives and protein drugs for cancer therapy. Nat Nanotechnol, 2014. 9(11): p. 907-912. Tan, S., Self-assembled Nanocomplexes Comprising of Green Tea Catechin Derivatives and Protein Drugs for Cancer Therapy. November 2013. Chen, I.J., et al., Therapeutic effect of high-dose green tea extract on weight reduction: A randomized, double-blind, placebo-controlled clinical trial. Clinical Nutrition, 2016. 35(3): p. 592-599. Zhang, S., et al., The Regulatory Effects and the Signaling Pathways of Natural Bioactive Compounds on Ferroptosis. Foods, 2021. 10(12): p. 2952. Chakrawarti, L., et al., Therapeutic effects of EGCG: a patent review. Expert Opin Ther Pat, 2016. 26(8): p. 907-16. Kose, T., et al., Curcumin and (-)- Epigallocatechin-3-Gallate Protect Murine MIN6 Pancreatic Beta-Cells Against Iron Toxicity and Erastin-Induced Ferroptosis. Pharmaceuticals (Basel, Switzerland), 2019. 12(1): p. 26. Rady, I., et al., Cancer preventive and therapeutic effects of EGCG, the major polyphenol in green tea. Egyptian Journal of Basic and Applied Sciences, 2018. 5(1): p. 1-23. Zhang, Y., et al., Global patterns and trends in ovarian cancer incidence: age, period and birth cohort analysis. BMC Cancer, 2019. 19(1): p. 984. Eisenhauer, E.A., Real-world evidence in the treatment of ovarian cancer. Annals of Oncology, 2017. 28: p. viii61-viii65. Bookman, M.A., et al., Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol, 2003. 21(2): p. 283-90. Wilken, J.A., K.T. Webster, and N.J. Maihle, Trastuzumab Sensitizes Ovarian Cancer Cells to EGFR-targeted Therapeutics. J Ovarian Res, 2010. 3: p. 7. Romond, E.H., et al., Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med, 2005. 353(16): p. 1673-84. Piccart-Gebhart, M.J., et al., Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Breast Cancer. New England Journal of Medicine, 2005. 353(16): p. 1659-1672. Joensuu, H., et al., Adjuvant Docetaxel or Vinorelbine with or without Trastuzumab for Breast Cancer. New England Journal of Medicine, 2006. 354(8): p. 809-820. Huang, Y.J., et al., Protective Effects of Epigallocatechin Gallate (EGCG) on Endometrial, Breast, and Ovarian Cancers. Biomolecules, 2020. 10(11). Peairs, A., et al., Epigallocatechin-3-gallate (EGCG) attenuates inflammation in MRL/lpr mouse mesangial cells. Cell Mol Immunol, 2010. 7(2): p. 123-32. Datta, S. and D. Sinha, EGCG maintained Nrf2-mediated redox homeostasis and minimized etoposide resistance in lung cancer cells. Journal of Functional Foods, 2019. 62: p. 103553. Li, C., et al., Green tea polyphenols modulate insulin secretion by inhibiting glutamate dehydrogenase. J Biol Chem, 2006. 281(15): p. 10214-21. Qing, G., et al., ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Cancer Cell, 2012. 22(5): p. 631-44. Choo, A.Y., et al., Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply. Mol Cell, 2010. 38(4): p. 487-99. Yang, C., et al., Glioblastoma cells require glutamate dehydrogenase to survive impairments of glucose metabolism or Akt signaling. Cancer Res, 2009. 69(20): p. 7986-93. J.M. Rabanel, V.A., I. Elkin, M. Mokhtar and P. Hildgen, Drug-Loaded Nanocarriers: Passive Targeting and Crossing of Biological Barriers. Current Medicinal Chemistry, 2012. 19, 3070–3102. Kim, S., et al., Overcoming the barriers in micellar drug delivery: loading efficiency, in vivo stability, and micelle-cell interaction. Expert Opin Drug Deliv, 2010. 7(1): p. 49-62. Rabanel, J.M., et al., Drug-loaded nanocarriers: passive targeting and crossing of biological barriers. Curr Med Chem, 2012. 19(19): p. 3070-102. Clynes, R.A., et al., Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med, 2000. 6(4): p. 443-6. Spiridon, C.I., S. Guinn, and E.S. Vitetta, A comparison of the in vitro and in vivo activities of IgG and F(ab')2 fragments of a mixture of three monoclonal anti-Her-2 antibodies. Clin Cancer Res, 2004. 10(10): p. 3542-51. Barok, M.r., et al., Trastuzumab causes antibody-dependent cellular cytotoxicity–mediated growth inhibition of submacroscopic JIMT-1 breast cancer xenografts despite intrinsic drug resistance. Molecular Cancer Therapeutics, 2007. 6(7): p. 2065-2072. | |
dc.identifier.uri | http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/84881 | - |
dc.description.abstract | 第二型人類表皮生長因子接受體(human epidermal growth factor receptor 2; HER2)屬於表皮生長因子家族的一員。過度表現HER2的癌細胞常與細胞不正常增生、腫瘤快速生長、存活率降低以及對常規化療藥物有較差的療效及預後有關。目前臨床上常以標靶藥物賀癌平(trastuzumab,商品名Herceptin)治療HER2過度表現的乳腺癌及胃癌,但是隨著療程時間增長,有限的藥物治療效果使得患者治療後期開始出現藥物療效大幅降低的現象。 本研究共分成兩個部分,其一為探討對賀癌平具有抗藥性的HER2陽性癌細胞之抗藥機制,其二為研究奈米載體聯合治療平台用於HER2陽性卵巢癌之影響。 根據臺灣衛福部統計數據指出女性乳癌為十大癌症中死亡年增率之冠,癌細胞對藥物產生抗藥性或腫瘤發生復發為其中可能原因之一,迫使癌症難以受到控制造成罹患乳癌婦女預後不良及死亡率增加,因此,了解對於HER2標靶治療產生抗藥性的癌細胞之潛在機制並且預防腫瘤復發的治療策略至關重要。近年來,鐵依賴性細胞死亡(ferroptosis)被認為是一種新的程序性死亡形式,由細胞膜中積累的脂質活性氧物質引起,有研究指出抗藥性癌細胞在藥物的作用下能夠躲避此死亡模式。 在本研究的第一部分中,我們發現在賀癌平與AKT抑制劑MK2206的合併治療下,會下調賀癌平阻抗的HER2陽性乳腺癌BT-474 clone 5細胞中的磷酸化AKT和GPX4的蛋白表達量,並增強抗藥性細胞對賀癌平的敏感度。此外,我們也發現利用ferroptosis的誘導劑erastin或是將細胞轉換至缺乏胱胺酸的環境能增強賀癌平對抗藥性HER2陽性乳腺癌BT-474 clone 5細胞的抑癌效果且有良好的藥效協同性,推測可能的機制是由於合併治療抑制了SLC7A11/GPX4信號通路並誘導鐵依賴性細胞死亡。最後將BT-474 clone 5細胞中的GPX4基因靜默(gene silencing)也觀察到有效恢復賀癌平抑癌效果。綜上所述,本研究提供了以下觀點:(1)PI3K/AKT信號通路與HER2過表達的賀癌平阻抗的乳腺癌有關;(2)改變CBS介導的轉硫化途徑不會影響HER2陽性乳腺癌細胞對賀癌平的抗藥性機制;(3)賀癌平會誘導癌細胞走向鐵依賴性細胞死亡途徑;(4)賀癌平阻抗的HER2陽性乳腺癌細胞透過調節AKT和SLC7A11的信號傳遞路徑以增強GPX4表達量來減緩癌細胞走向鐵依賴性細胞死亡,侷限賀癌平對癌細胞的抑制作用。 卵巢癌為常見且致命的婦科癌症,儘管已經開發出許多新的治療方針,但患者的存活率仍然很低。根據統計指出卵巢癌病中HER2呈現陽性的占比約20%,雖然賀癌平已被FDA批准用於治療HER2陽性的乳腺癌和胃癌病患,但應用於卵巢癌病患中卻不見顯著療效。 綠茶萃取物中最豐富的多酚成分是表沒食子兒茶素沒食子酸酯(EGCG),多項研究顯示其功能在對抗心血管疾病、神經退行性疾病、糖尿病和癌症等方面表現皆具有良好的生物活性,因此近年來,人們關注開發EGCG參與的癌症靶向治療的抗癌策略。微胞奈米複合物(micellar nanocomplex)的設計核心是利用低聚合EGCG(Oligomerized EGCG, OEGCG)與抗癌蛋白藥物賀癌平自組裝形成,再將聚乙二醇修飾之EGCG(PEG-EGCG)複合形成外殼,全組合成新型奈米載體聯合療法並將之稱作nano-trastuzumab。利用此藥物靜脈注射至卵巢癌(SKOV3-Luc)小鼠異種移植模型中,給予EGCG作為載體並裝載賀癌平的nano-trastuzumab之組別顯示出比單賀癌平使用有更好的抑癌效果並有效抑制腫瘤生長,因此,在本研究的第二個部分中,發現奈米載體聯合療法nano-trastuzumab有望成為HER2陽性卵巢癌細胞的一個新興治療策略。 | zh_TW |
dc.description.abstract | Human epidermal growth factor receptor 2 (HER2) belongs to the epidermal growth factor receptor family of proto-oncogenes. Its amplification is associated with rapid tumor growth, shortened survival, and poor response to conventional chemotherapeutic agents. The HER2-targeting antibody trastuzumab has shown effectiveness in treating HER2-positive breast and gastric cancers; however, its responses are limited. This study is divided into two parts. The first part is to investigate the molecular mechanisms of resistance in HER2-positive cancer treated with trastuzumab. The other is to investigate the anticancer effects of nanocarrier combination therapy platforms in ovarian cancer overexpressing HER2. Drug resistance makes it difficult for cancer therapies to achieve stable and complete responses. Emerging evidence suggests that amino acid reprogramming leads to cancer cells resistant to conventional chemotherapeutic drugs and plays an important role in the survival of residual drug-resistant cells. Targeting resistant cells thus provides a therapeutic opportunity to prevent tumor recurrence. Ferroptosis is a type of iron-dependent cell death caused by the accumulation of lipid reactive oxygen species in cell membranes. This new form of programmed cell death might play a role in drug resistance mechanisms. In the first part of this study, we reported that combining MK2206, an AKT inhibitor, with trastuzumab exerted the growth-inhibitory effects on trastuzumab-resistant HER2-positive breast cancer BT-474 clone 5 cells by dampening AKT and GPX4 signaling pathways. Moreover, We found that erastin, a ferroptosis inducer by inhibiting xCT (encoded by SLC7A11), or cystine deprivation enhanced the anticancer effects of trastuzumab on BT-474 clone 5 cells, which may be attributed to the upregulation of AKT/SLC7A11/GPX4 axis inhibited ferroptosis pathway and further conferred trastuzumab resistance in BT-474 clone 5 cells. Furthermore, silencing GPX4 expression enhanced the sensitivity to trastuzumab in trastuzumab-resistant HER2-positive breast cancer BT-474 clone 5 cells. In conclusion, the study provided insights into (1) PI3K/AKT pathway was involved in the resistance to trastuzumab in HER2-positive breast cancer; (2) CBS-mediated transsulfuration pathway may not be involved in HER2-positive breast cancer resistance to trastuzumab; (3) trastuzumab reduced GPX4 protein expression and induced ferroptosis; (4) mechanistic studies demonstrated that trastuzumab-resistant HER2-positive breast cancer cells prevent ferroptosis by regulating AKT and SLC7A11/GPX4 signaling pathway and enhancing GPX4 expression. The second part looked into how the nanocarrier combination therapy platform affected ovarian cancer that overexpressed HER2 at a rate similar to many malignant breast tumors. Ovarian cancer (OC) is a common and deadly gynecological cancer. Although new therapeutic agents for OC have been developed, patient survival remains low. Trastuzumab is approved for breast and gastric cancers with high HER2 expression, but it has not shown clinical success in OC. The most abundant polyphenolic constituent from green tea extract is epigallocatechin gallate (EGCG), which has demonstrated versatile bioactivities in combating cardiovascular diseases, neurodegenerative diseases, diabetes, and cancer. Because of its anticancer activity, there has been an increase in interest in developing effective strategies involving EGCG in cancer target therapy. The core of the micellar nanocomplex is formed by complexing oligomerized EGCG (OEGCG) with trastuzumab, followed by complexation of poly (ethylene glycol)-EGCG (PEG-EGCG) to form the shell, which is named nano-trastuzumab. The nano-trastuzumab showed better growth reduction than free trastuzumab when injected into the ovarian (SKOV3-Luc) xenograft mouse model. As a result, in the second part, we demonstrated that nano-trastuzumab could be a promising treatment for HER2-positive ovarian cancer. | en |
dc.description.provenance | Made available in DSpace on 2023-03-19T22:30:36Z (GMT). No. of bitstreams: 1 U0001-2608202213051700.pdf: 3433693 bytes, checksum: 573cfcb150ac8b5cc239b67dfe2ba25f (MD5) Previous issue date: 2022 | en |
dc.description.tableofcontents | 口試委員會審定書 i 誌謝 ii 中文摘要 iii Abstract v List of Abbreviations viii List of Figures xiv Section 1–Identification of resistance mechanism associated with trastuzumab resistance in HER2-positive breast cancer cell lines 1 Chapter 1 –Introduction 1 1.1 HER family 1 1.2 HER receptors in carcinogenesis 2 1.3 HER2-amplified cancers 4 1.4 Approved indications for HER2 targeted therapy 5 Trastuzumab 5 1.5 Acquired therapeutic resistance 6 1.6 Metabolic reprogramming 8 1.6.1. Regulation of glucose metabolism in cancer cells 8 1.6.2. Regulation of amino acid metabolism in cancer cells 9 1.6.3. Regulation of fatty acid metabolism in cancer cells 12 Chapter 2 –Specific aim 16 Chapter 3 –Materials and Methods 17 3.1 Drugs 17 3.2 Cell culture 17 3.3 Cell viability assay 18 3.4 Combination effect analysis 19 3.5 Western blotting 19 3.6 Immunofluorescence staining 20 3.7 Transwell cell migration assay 21 3.8 Bioinformatics analysis 21 3.9 Lipid peroxidation assay 22 3.10 Lentivirus production and transient transduction 22 3.11 Statistical analysis 23 Chapter 4 -Results 24 4.1 Trastuzumab resistance and HER2 status were confirmed in BT-474 cell lines. 24 4.2 BT-474 clone 5 exhibited migratory and mesenchymal phenotypes. 25 4.3 PI3K/AKT Pathway was reprogrammed in BT-474 clone 5 25 4.4 Cysteine and methionine metabolism pathways were altered in the trastuzumab-resistant breast cancer cell line 27 4.5 BT-474 clone 5 cells might express higher CBS for synthesizing intracellular cysteine to survive from cystine deprivation 28 4.6 CBS might not be a prominent modulator of trastuzumab resistance 29 4.7 Trastuzumab-induced cell death was accelerated in the absence of extracellular cysteine uptake 30 4.8 Higher GPX4 expression may protect BT-474 clone 5 cells from trastuzumab-induced ferroptosis 30 4.9 The upregulation of AKT/SLC7A11/GPX4 axis inhibited ferroptosis pathway and further conferred trastuzumab resistance in BT-474 clone 5 cells 31 4.10 Silencing expression of GPX4 reversed trastuzumab resistance 33 4.11 Correlation between GPX4 expression and the survival of HER2-positive breast cancer patients 33 Chapter 5 –Conclusion 35 Chapter 6 -Discussion 36 Chapter 7 –Figures 41 Section 2 –Efficacy testing of a nanocarrier combination therapy platform for HER2-positive cancers 56 Chapter 1-Introduction 56 1.1 Nanotechnology in drug delivery for cancer therapy 56 1.2 Enhanced permeability and retention effect 57 1.3 Current nanotechnology therapeutics delivery platforms 58 1.3.1. Liposomes 59 1.3.2 Polymeric micelles 60 1.3.3 Dendrimers 60 1.3.4 Metal nanoparticles 61 1.3.5 Nanoparticle albumin-bound (nab) 62 1.4 Nanocarriers used in drug delivery system 62 1.5 Design of “Nano-trastuzumab” for cancer therapy 64 1.6 Green tea catechins 65 Chapter 2 –Specific aim 67 Chapter 3 –Materials and Methods 68 3.1 Drugs 68 3.2 Cell culture 68 3.3 In vitro cell viability 69 3.4 Lentivirus production and stable transduction 69 3.5 Luciferase assay 70 3.6 Animal model 70 3.7 Drug treatment 71 3.8 In vivo BLI of orthotopic ovarian cancer models during drug treatment 71 3.9 Body weight 71 3.10 Statistical analysis 72 Chapter 4 -Results 73 4.1 The EGCG-based nanocomplex inhibited ovarian cancer cell viability while having minimal cytotoxicity in normal cells. 73 4.2 Inhibition of in vivo tumor growth by nano-trastuzumab in the early stage 73 4.3 Tumors began to recur after drug withdrawal over a long time 74 4.4 Macroscopic observation of ovarian intraperitoneal xenografts in nude mice 75 Chapter 5 –Conclusion 76 Chapter 6 -Discussion 77 Chapter 7 –Figures 81 Chapter 8 –References 87 | |
dc.language.iso | en | |
dc.title | 探討第二型人類表皮生長因子受體陽性癌症之抗藥性機制 | zh_TW |
dc.title | Investigating resistance mechanisms to anti-HER2 therapy for HER2-positive cancers | en |
dc.type | Thesis | |
dc.date.schoolyear | 110-2 | |
dc.description.degree | 碩士 | |
dc.contributor.oralexamcommittee | 林亮音(Liang-In Lin),楊雅倩(Ya-Chien Yang),蘇剛毅(Kang-Yi Su) | |
dc.subject.keyword | 第二型人類上皮生長因子接受體,賀癌平,抗藥性,胱胺酸和甲硫胺酸代謝途徑,鐵依賴性死亡,穀胱甘肽過氧化物酶,奈米藥物載體,表沒食子兒茶素沒食子酸酯, | zh_TW |
dc.subject.keyword | HER2,trastuzumab,drug resistance,cysteine and methionine metabolism,ferroptosis,GPX4,EGCG,nanocarrier,micellar nanocomplex, | en |
dc.relation.page | 97 | |
dc.identifier.doi | 10.6342/NTU202202849 | |
dc.rights.note | 同意授權(限校園內公開) | |
dc.date.accepted | 2022-08-29 | |
dc.contributor.author-college | 醫學院 | zh_TW |
dc.contributor.author-dept | 醫學檢驗暨生物技術學研究所 | zh_TW |
dc.date.embargo-lift | 2022-10-05 | - |
顯示於系所單位: | 醫學檢驗暨生物技術學系 |
文件中的檔案:
檔案 | 大小 | 格式 | |
---|---|---|---|
U0001-2608202213051700.pdf 授權僅限NTU校內IP使用(校園外請利用VPN校外連線服務) | 3.35 MB | Adobe PDF | 檢視/開啟 |
系統中的文件,除了特別指名其著作權條款之外,均受到著作權保護,並且保留所有的權利。