| 研究生: |
林靜渝 Lin, Ching-Yu |
|---|---|
| 論文名稱: |
以具酸鹼/氧化還原敏感之聚胺酸類微胞包覆阿黴素運用於癌症治療:體外及體內評估 Redox/pH Dual-Responsive Polypeptide Assemblies to Enhance the Antitumor Efficacy of Doxorubicin: In vitro and In vivo Evaluation |
| 指導教授: |
葉明龍
Yeh, Ming-Long |
| 共同指導教授: |
詹正雄
Jan, Jeng-Shiung |
| 學位類別: |
碩士 Master |
| 系所名稱: |
工學院 - 生物醫學工程學系 Department of BioMedical Engineering |
| 論文出版年: | 2016 |
| 畢業學年度: | 105 |
| 語文別: | 英文 |
| 論文頁數: | 78 |
| 中文關鍵詞: | 聚賴胺酸 、聚酪胺酸 、環境刺激 、藥物傳遞 、阿黴素 、癌症治療 |
| 外文關鍵詞: | polylysine, polytyrosine, stimuli-responsive, drug delivery, doxorubicin, cancer treatment |
| 相關次數: | 點閱:76 下載:2 |
| 分享至: |
| 查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報 |
化學療法被廣泛運用於治療癌症,但注射化療藥物後藥物沒有專一性地分佈於體內造成副作用。因此,可以利用藥物傳遞系統增加藥物的專一性以及藥物在腫瘤處的累積量以達到更良好的治療效果。聚胺酸類的高分子可在溶液中自組裝因為其具有特殊的二級結構,當其裝載藥物時,可藉由調控外界刺激控制藥物的釋放。本研究合成聚乙二醇-聚賴胺酸-聚酪胺酸高分子,並利用DTSSP交聯劑引入雙硫鍵,形成具酸鹼/氧化還原敏感之聚胺酸類微胞。利用此微胞包覆阿黴素形成粒徑約63奈米且載藥率約為16.5%的載體。利用穿透式電子顯微鏡觀察發現此微胞在pH 7.4的環境下呈現均一的球形,然而已交聯的微胞在模擬腫瘤組織的酸性(pH 5.0)以及具高氧化還原物質(GSH 10mM)的環境中呈現不規則聚集且降解的情形。體外實驗中發現,對於路易士肺癌細胞未利用載體所包覆的阿黴素相較於載藥之微胞具有較高的細胞毒性,但載藥之微胞可以減低DOX對於纖維母細胞的毒性。此外,在增生實驗中發現此載藥之微胞能有效且選擇性抑制路易士肺癌細胞的增生,此現象在模擬腫瘤環境的酸性培養液中更為顯著。體內實驗中在皮下路易士腫瘤小鼠模型中,證實此載藥之微胞比起未利用載體所包覆的阿黴素更具腫瘤抑制效果並且降低藥物的副作用,進一步證實利用聚乙二醇-聚賴胺酸-聚酪胺酸高分子包覆阿黴素有利於增進化療藥物的療效,在癌症治療應用上具有潛力。
Nanocarriers are promising to benefit cancer therapy. Stimuli-responsive polypeptide-based copolymers can self-assemble in solution, and release their payloads under specific stimuli due to their specific secondary structure. To enhance site-specific drug release in tumor tissues and the antitumor efficacy of antitumor agents, pH/redox-responsive poly(ethylene glycol)-b-poly(L-lysine-co-L-tyrosine) copolymer (PEG-b-(Lys-co-Tyr)) was developed to encapsulate doxorubicin (DOX) and cross-link with 3,3’-dithiobis(sulfosuccinimidylpropionate) (DTSSP). The DOX-loaded assemblies had diameters around 63 nm and a superior drug-loading content (DLC) (~16.5%). Under TEM observation, polypeptide assemblies showed uniformly spherical morphologies in pH 7.4 environment; however, cross-linked polypeptide assemblies demonstrated redox-triggered degradation and amorphous aggregates in pH 5.0 and GSH (10 mM) conditions. Cytotoxicity studies showed that free DOX was more toxic than DOX-loaded assemblies against LLC cells; however, DOX-loaded assemblies decreased the burden of DOX on NIH3T3 cells. The cell uptake level of free DOX was slightly higher than that of DOX-loaded assemblies. Proliferation assay showed that DOX-loaded assemblies can selectively inhibit Lewis lung carcinoma (LLC) cells proliferation especially in acidic environment. Moreover, in vivo studies validated that DOX-loaded assemblies showed significant antitumor efficacy on LLC cells subcutaneous model and reduced the side effects, indicating its great potential for cancer treatments.
[1] Bruce Alberts AJ, Julian Lewis, Martin Raff, Keith Roberts, and Peter Walter. Molecular Biology of the Cell 4th edn. Garland Science. New York. (2002).
[2] Siegel RL, Miller KD, et al. Cancer Statistics, 2015. CA: A Cancer Journal for Clinicians. 65 (1): 5-29 (2015).
[3] Fitzmaurice C, Dicker D, et al. The Global Burden of Cancer 2013. JAMA Oncology. 1 (4): 505-27 (2015).
[4] American Cancer Society. Key statistics for lung cancer. (2016).
[5] Travis WD. Pathology of lung cancer. Clinics in Chest Medicine. 32 (4): 669-92 (2011).
[6] Alberg AJ, Samet JM. Epidemiology of lung cancer. Chest. 123 (1 Suppl): 21s-49s (2003).
[7] Lorigan P, Radford J, et al. Lung cancer after treatment for Hodgkin's lymphoma: a systematic review. The Lancet Oncology. 6 (10): 773-9 (2005).
[8] Kaufman EL, Jacobson JS, et al. Effect of breast cancer radiotherapy and cigarette smoking on risk of second primary lung cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 26 (3): 392-8 (2008).
[9] Fontham ET, Correa P, et al. Environmental tobacco smoke and lung cancer in nonsmoking women. A multicenter study. JAMA Oncology. 271 (22): 1752-9 (1994).
[10] Aberle DR, Adams AM, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. The New England Journal of Medicine. 365 (5): 395-409 (2011).
[11] Woolf SH, Harris RP, et al. Low-dose computed tomography screening for lung cancer: how strong is the evidence? JAMA Internal Medicine. 174 (12): 2019-22 (2014).
[12] Hoffman PC, Mauer AM, et al. Lung cancer. Lancet. 355 (9202): 479-85 (2000).
[13] Manser R, Wright G, et al. Surgery for early stage non-small cell lung cancer. The Cochrane Database of Systematic Reviews. (1): Cd004699 (2005).
[14] Gajra A, Newman N, et al. Effect of number of lymph nodes sampled on outcome in patients with stage I non-small-cell lung cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 21 (6): 1029-34 (2003).
[15] Wang EH, Corso CD, et al. Postoperative Radiation Therapy Is Associated With Improved Overall Survival in Incompletely Resected Stage II and III Non-Small-Cell Lung Cancer. Journal of Clinical Oncology : official journal of the American Society of Clinical Oncology. 33 (25): 2727-34 (2015).
[16] Trodella L, Granone P, et al. Adjuvant radiotherapy in non-small cell lung cancer with pathological stage I: definitive results of a phase III randomized trial. Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology. 62 (1): 11-9 (2002).
[17] Thomas M, Rube C, et al. Effect of preoperative chemoradiation in addition to preoperative chemotherapy: a randomised trial in stage III non-small-cell lung cancer. The Lancet Oncology. 9 (7): 636-48 (2008).
[18] Gaspar LE, Gay EG, et al. Limited-stage small-cell lung cancer (stages I-III): observations from the National Cancer Data Base. Clinical Lung Cancer. 6 (6): 355-60 (2005).
[19] Bissell MJ, Hines WC. Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nature Medicine. 17 (3): 320-9 (2011).
[20] Ronnov-Jessen L, Petersen OW. Induction of alpha-smooth muscle actin by transforming growth factor-beta 1 in quiescent human breast gland fibroblasts. Implications for myofibroblast generation in breast neoplasia. Laboratory Investigation. 68 (6): 696-707 (1993).
[21] Olumi AF, Grossfeld GD, et al. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Research. 59 (19): 5002-11 (1999).
[22] Nguyen-Ngoc K-V, Cheung KJ, et al. ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proceedings of the National Academy of Sciences. 109 (39): E2595–E604 (2012).
[23] Khamis ZI, Sahab ZJ, et al. Active roles of tumor stroma in breast cancer metastasis. International Journal of Breast Cancer. 2012: 574025 (2012).
[24] Kessenbrock K, Plaks V, et al. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 141 (1): 52-67 (2010).
[25] Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radical Biology & Medicine. 30 (11): 1191-212 (2001).
[26] McCarley RL. Redox-Responsive Delivery Systems. Annual Review of Analytical Chemistry. 5: 391-411 (2012).
[27] Balendiran GK, Dabur R, et al. The role of glutathione in cancer. Cell Biochemistry and Function. 22 (6): 343-52 (2004).
[28] Manickam DS, Li J, et al. Effect of innate glutathione levels on activity of redox-responsive gene delivery vectors. Journal of Controlled Release. 141 (1): 77-84 (2010).
[29] Ortega AL, Mena S, et al. Oxidative and nitrosative stress in the metastatic microenvironment. Cancers. 2 (2): 274-304 (2010).
[30] Ames BN, Gold LS, et al. The causes and prevention of cancer. Proceedings of the National Academy of Sciences of the United States of America. 92 (12): 5258-65 (1995).
[31] Franco R, Sanchez-Olea R, et al. Environmental toxicity, oxidative stress and apoptosis: menage a trois. Mutation Research. 674 (1-2): 3-22 (2009).
[32] Terradez P, Asensi M, et al. Depletion of tumour glutathione in vivo by buthionine sulphoximine: modulation by the rate of cellular proliferation and inhibition of cancer growth. The Biochemical Journal. 292 (Pt 2): 477-83 (1993).
[33] Suzuki M, Takahashi T, et al. Medial regression and its functional significance in tumor-supplying host arteries. A morphometric study of hepatic arteries in human livers with hepatocellular carcinoma. Cancer. 59 (3): 444-50 (1987).
[34] Skinner SA, Tutton PJ, et al. Microvascular architecture of experimental colon tumors in the rat. Cancer Research. 50 (8): 2411-7 (1990).
[35] Seymour LW, Miyamoto Y, et al. Influence of molecular weight on passive tumour accumulation of a soluble macromolecular drug carrier. European Journal of Cancer. 31A (5): 766-70 (1995).
[36] Folkman J. Angiogenesis: an organizing principle for drug discovery? Nature Reviews Drug Discovery. 6 (4): 273-86 (2007).
[37] Jain RK, Wardhartley K. Tumor Blood-Flow - Characterization, Modifications, and Role in Hyperthermia. IEEE Transactions on Sonics and Ultrasonics. 31 (5): 504-26 (1984).
[38] Eddy HA. Microangiographic techniques in the study of normal and tumor tissue vascular systems. Microvascular Research. 11 (3): 391-413 (1976).
[39] Svastova E, Hulikova A, et al. Hypoxia activates the capacity of tumor-associated carbonic anhydrase IX to acidify extracellular pH. FEBS Letters. 577 (3): 439-45 (2004).
[40] Webb SD, Sherratt JA, et al. Mathematical modelling of tumour acidity: regulation of intracellular pH. Journal of Theoretical Biology. 196 (2): 237-50 (1999).
[41] Delbaldo C, Michiels S, et al. Benefits of adding a drug to a single-agent or a 2-agent chemotherapy regimen in advanced non-small-cell lung cancer: a meta-analysis. JAMA. 292 (4): 470-84 (2004).
[42] Campagna M-C. Chemotherapy Complications. Hospital Medicine Clinics. 5 (3): 400-12 (2016).
[43] Lomovskaya N, Otten SL, et al. Doxorubicin Overproduction in Streptomyces peucetius: Cloning and Characterization of the dnrU Ketoreductase and dnrV Genes and the doxA Cytochrome P-450 Hydroxylase Gene. Journal of Bacteriology. 181 (1): 305-18 (1999).
[44] Tacar O, Sriamornsak P, et al. Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. Journal of Pharmacy and Pharmacology. 65 (2): 157-70 (2013).
[45] Minotti G, Menna P, et al. Anthracyclines: Molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacological Reviews. 56 (2): 185-229 (2004).
[46] Chen NT, Wu CY, et al. Probing the Dynamics of Doxorubicin-DNA Intercalation during the Initial Activation of Apoptosis by Fluorescence Lifetime Imaging Microscopy (FLIM). PLoS One. 7 (9): (2012).
[47] Aubelsadron G, Londosgagliardi D. Daunorubicin and Doxorubicin, Anthracycline Antibiotics, a Physicochemical and Biological Review. Biochimie. 66 (5): 333-52 (1984).
[48] Mizutani H, Tada-Oikawa S, et al. Mechanism of apoptosis induced by doxorubicin through the generation of hydrogen peroxide. Life Sciences. 76 (13): 1439-53 (2005).
[49] Nitiss JL. Investigating the biological functions of DNA topoisomerases in eukaryotic cells. Biochimica Et Biophysica Acta-Gene Structure and Expression. 1400 (1-3): 63-81 (1998).
[50] Tewey KM, Rowe TC, et al. Adriamycin-Induced DNA Damage Mediated by Mammalian DNA Topoisomerase-Ii. Science. 226 (4673): 466-8 (1984).
[51] Bates DA, Winterbourn CC. Deoxyribose breakdown by the adriamycin semiquinone and H2O2: evidence for hydroxyl radical participation. FEBS Letters. 145 (1): 137-42 (1982).
[52] Di X, Shiu RP, et al. Apoptosis, autophagy, accelerated senescence and reactive oxygen in the response of human breast tumor cells to adriamycin. Biochemical Pharmacology. 77 (7): 1139-50 (2009).
[53] Skladanowski A, Konopa J. Adriamycin and daunomycin induce programmed cell death (apoptosis) in tumour cells. Biochemical Pharmacology. 46 (3): 375-82 (1993).
[54] Lockshin RA, Zakeri Z. Apoptosis, autophagy, and more. The International Journal of Biochemistry & Cell Biology. 36 (12): 2405-19 (2004).
[55] Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Current Opinion in Cell Biology. 16 (6): 663-9 (2004).
[56] Hsieh YC, Athar M, et al. When apoptosis meets autophagy: deciding cell fate after trauma and sepsis. Trends in Molecular Medicine. 15 (3): 129-38 (2009).
[57] Zhang YW, Shi J, et al. Cardiomyocyte death in doxorubicin-induced cardiotoxicity. Archivum Immunologiae et Therapiae Experimentalis. 57 (6): 435-45 (2009).
[58] Zhou YY, Li Y, et al. MAPK/JNK signalling: a potential autophagy regulation pathway. Bioscience Reports. 35 (3): (2015).
[59] Ashkenazi A. Directing cancer cells to self-destruct with pro-apoptotic receptor agonists. Nature Reviews Drug Discovery. 7 (12): 1001-12 (2008).
[60] Smuder AJ, Kavazis AN, et al. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. Journal of Applied Physiology. 111 (4): 1190-8 (2011).
[61] Czarny P, Pawlowska E, et al. Autophagy in DNA damage response. International Journal of Molecular Sciences. 16 (2): 2641-62 (2015).
[62] Shin HJ, Kwon HK, et al. Doxorubicin-induced necrosis is mediated by poly-(ADP-ribose) polymerase 1 (PARP1) but is independent of p53. Scientific Reports. 5: 15798 (2015).
[63] Zong WX, Ditsworth D, et al. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes & Development. 18 (11): 1272-82 (2004).
[64] U.S. National Institutes of Health. (2015).
[65] Venturoli D, Rippe B. Ficoll and dextran vs. globular proteins as probes for testing glomerular permselectivity: effects of molecular size, shape, charge, and deformability. American Journal of Physiology - Renal Physiology. 288 (4): F605-F13 (2005).
[66] Aderem A, Underhill DM. Mechanisms of phagocytosis in macrophages. Annual Review of Immunology. 17: 593-623 (1999).
[67] Wisse E, Braet F, et al. Structure and function of sinusoidal lining cells in the liver. Toxicologic Pathology. 24 (1): 100-11 (1996).
[68] Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nature Reviews Cancer. 6 (8): 583-92 (2006).
[69] Harris JM, Martin NE, et al. Pegylation: a novel process for modifying pharmacokinetics. Clinical Pharmacokinetics. 40 (7): 539-51 (2001).
[70] Yamaoka T, Tabata Y, et al. Distribution and tissue uptake of poly(ethylene glycol) with different molecular weights after intravenous administration to mice. Journal of Pharmaceutical Sciences. 83 (4): 601-6 (1994).
[71] Chen MY, Hoffer A, et al. Surface properties, more than size, limiting convective distribution of virus-sized particles and viruses in the central nervous system. Journal of Neurosurgery. 103 (2): 311-9 (2005).
[72] Maeda H, Ueda M, et al. Conjugation of poly(styrene-co-maleic acid) derivatives to the antitumor protein neocarzinostatin: pronounced improvements in pharmacological properties. Journal of Medicinal Chemistry. 28 (4): 455-61 (1985).
[73] Greish K. Enhanced permeability and retention of macromolecular drugs in solid tumors: a royal gate for targeted anticancer nanomedicines. Journal of Drug Targeting. 15 (7-8): 457-64 (2007).
[74] Beduneau A, Saulnier P, et al. Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab' fragments. Biomaterials. 28 (33): 4978-90 (2007).
[75] Stuart MAC, Huck WTS, et al. Emerging applications of stimuli-responsive polymer materials. Nature Materials. 9 (2): 101-13 (2010).
[76] Wu H, Zhu L, et al. pH-sensitive poly(histidine)-PEG/DSPE-PEG co-polymer micelles for cytosolic drug delivery. Biomaterials. 34 (4): 1213-22 (2013).
[77] Li G, Meng Y, et al. Formation of thermo-sensitive polyelectrolyte complex micelles from two biocompatible graft copolymers for drug delivery. Journal of Biomedical Materials Research Part A. 102 (7): 2163-72 (2014).
[78] Wang AZ, Langer R, et al. Nanoparticle Delivery of Cancer Drugs. Annual Review of Medicine. 63 (1): 185-98 (2012).
[79] Zhao Y, Butler EB, et al. Targeting cellular metabolism to improve cancer therapeutics. Cell Death & Disease. 4: e532 (2013).
[80] Duncan R, Gaspar R. Nanomedicine(s) under the Microscope. Molecular Pharmaceutics. 8 (6): 2101-41 (2011).
[81] Huang HC, Barua S, et al. Inorganic nanoparticles for cancer imaging and therapy. Journal of Controlled Release. 155 (3): 344-57 (2011).
[82] Duncan R. Polymer conjugates as anticancer nanomedicines. Nature Reviews Cancer. 6 (9): 688-701 (2006).
[83] Seymour LW, Ferry DR, et al. Hepatic Drug Targeting: Phase I Evaluation of Polymer-Bound Doxorubicin. Journal of Clinical Oncology. 20 (6): 1668-76 (2002).
[84] Barenholz Y. Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release. 160 (2): 117-34 (2012).
[85] Chan S, Davidson N, et al. Phase III trial of liposomal doxorubicin and cyclophosphamide compared with epirubicin and cyclophosphamide as first-line therapy for metastatic breast cancer. Annals of Oncology. 15 (10): 1527-34 (2004).
[86] Gill PS, Wernz J, et al. Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi's sarcoma. Journal of Clinical Oncology. 14 (8): 2353-64 (1996).
[87] Gokbuget N, Hartog CM, et al. Liposomal cytarabine is effective and tolerable in the treatment of central nervous system relapse of acute lymphoblastic leukemia and very aggressive lymphoma. Haematologica. 96 (2): 238-44 (2011).
[88] O'Brien S, Schiller G, et al. High-dose vincristine sulfate liposome injection for advanced, relapsed, and refractory adult Philadelphia chromosome-negative acute lymphoblastic leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 31 (6): 676-83 (2013).
[89] Frampton JE. Mifamurtide. Pediatric Drugs. 12 (3): 141-53 (2010).
[90] Wicki A, Witzigmann D, et al. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. Journal of Controlled Release. 200: 138-57 (2015).
[91] Wicki A, Rochlitz C, et al. Targeting Tumor-Associated Endothelial Cells: Anti-VEGFR2 Immunoliposomes Mediate Tumor Vessel Disruption and Inhibit Tumor Growth. American Association for Cancer Research. 18 (2): 454-64 (2012).
[92] Huwyler J, Drewe J, et al. Tumor targeting using liposomal antineoplastic drugs. International Journal of Nanomedicine. 3 (1): 21-9 (2008).
[93] Kamaly N, Xiao Z, et al. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chemical Society Reviews. 41 (7): 2971-3010 (2012).
[94] Von Hoff DD, Mita MM, et al. Phase I Study of PSMA-Targeted Docetaxel-Containing Nanoparticle BIND-014 in Patients with Advanced Solid Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 22 (13): 3157-63 (2016).
[95] Rijcken CJ, Snel CJ, et al. Hydrolysable core-crosslinked thermosensitive polymeric micelles: Synthesis, characterisation and in vivo studies. Biomaterials. 28 (36): 5581-93 (2007).
[96] Desai N, Trieu V, et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. American Association for Cancer Research. 12 (4): 1317-24 (2006).
[97] Qu W, Chen W-H, et al. Avidin–Biotin Interaction Mediated Peptide Assemblies as Efficient Gene Delivery Vectors for Cancer Therapy. Molecular Pharmaceutics. 10 (1): 261-9 (2013).
[98] Kricheldorf HR. Polypeptides and 100 years of chemistry of alpha-amino acid N-carboxyanhydrides. Angewandte Chemie International Edition. 45 (35): 5752-84 (2006).
[99] Deming TJ. Synthetic polypeptides for biomedical applications. Progress in Polymer Science. 32 (8-9): 858-75 (2007).
[100] Bae Y, Kataoka K. Intelligent polymeric micelles from functional poly(ethylene glycol)-poly(amino acid) block copolymers. Advanced Drug Delivery Reviews. 61 (10): 768-84 (2009).
[101] Lv S, Li M, et al. Doxorubicin-loaded amphiphilic polypeptide-based nanoparticles as an efficient drug delivery system for cancer therapy. Acta Biomaterialia. 9 (12): 9330-42 (2013).
[102] O'Reilly RK, Hawker CJ, et al. Cross-linked block copolymer micelles: functional nanostructures of great potential and versatility. Chemical Society Reviews. 35 (11): 1068-83 (2006).
[103] Koo AN, Lee HJ, et al. Disulfide-cross-linked PEG-poly(amino acid)s copolymer micelles for glutathione-mediated intracellular drug delivery. Chemical Communications. (48): 6570-2 (2008).
[104] Briza P, Ellinger A, et al. Characterization of a Dl-Dityrosine-Containing Macromolecule from Yeast Ascospore Walls. Journal of Biological Chemistry. 265 (25): 15118-23 (1990).
[105] Huang YF, Lu SC, et al. Cross-Linked, Self-Fluorescent Gold Nanoparticle/Polypeptide Nanocapsules Comprising Dityrosine for Protein Encapsulation and Label-Free Imaging. Small. 10 (10): 1939-44 (2014).
[106] Younes A, Gopal AK, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin's lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 30 (18): 2183-9 (2012).
[107] Burris HA, 3rd, Rugo HS, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 29 (4): 398-405 (2011).
[108] Rivera E. Liposomal anthracyclines in metastatic breast cancer: clinical update. The Oncologist. 8 (Suppl 2): 3-9 (2003).
[109] Glantz MJ, Jaeckle KA, et al. A randomized controlled trial comparing intrathecal sustained-release cytarabine (DepoCyt) to intrathecal methotrexate in patients with neoplastic meningitis from solid tumors. Clinical Cancer Research : an Official Journal of the American Association for Cancer Research. 5 (11): 3394-402 (1999).
[110] Muggia FM, Hainsworth JD, et al. Phase II study of liposomal doxorubicin in refractory ovarian cancer: antitumor activity and toxicity modification by liposomal encapsulation. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology. 15 (3): 987-93 (1997).
[111] Lee KS, Chung HC, et al. Multicenter phase II trial of Genexol-PM, a Cremophor-free, polymeric micelle formulation of paclitaxel, in patients with metastatic breast cancer. Breast Cancer Research and Treatment. 108 (2): 241-50 (2008).
[112] Khemapech N, Oranratanaphan S, et al. Salvage chemotherapy in recurrent platinum-resistant or refractory epithelial ovarian cancer with Carboplatin and distearoylphosphatidylcholine pegylated liposomal Doxorubicin (lipo-dox(R)). Asian Pacific Journal of Cancer Prevention. 14 (3): 2131-5 (2013).
[113] Venkatakrishnan K, Liu Y, et al. Pharmacokinetics and pharmacodynamics of liposomal mifamurtide in adult volunteers with mild or moderate hepatic impairment. British Journal of Clinical Pharmacology. 77 (6): 998-1010 (2014).
[114] Batist G, Ramakrishnan G, et al. Reduced cardiotoxicity and preserved antitumor efficacy of liposome-encapsulated doxorubicin and cyclophosphamide compared with conventional doxorubicin and cyclophosphamide in a randomized, multicenter trial of metastatic breast cancer. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology. 19 (5): 1444-54 (2001).
[115] Rodriguez MA, Pytlik R, et al. Vincristine sulfate liposomes injection (Marqibo) in heavily pretreated patients with refractory aggressive non-Hodgkin lymphoma: report of the pivotal phase 2 study. Cancer. 115 (15): 3475-82 (2009).
[116] Dinndorf PA, Gootenberg J, et al. FDA Drug Approval Summary: Pegaspargase (Oncaspar®) for the First-Line Treatment of Children with Acute Lymphoblastic Leukemia (ALL). The Oncologist. 12 (8): 991-8 (2007).
[117] Maeda H. SMANCS and polymer-conjugated macromolecular drugs: advantages in cancer chemotherapy. Advanced Drug Delivery Reviews. 46 (1–3): 169-85 (2001).
[118] Danhauser-Riedl S, Hausmann E, et al. Phase I clinical and pharmacokinetic trial of dextran conjugated doxorubicin (AD-70, DOX-OXD). Investigational New Drugs. 11 (2-3): 187-95 (1993).
[119] Vasey PA, Kaye SB, et al. Phase I clinical and pharmacokinetic study of PK1 [N-(2-hydroxypropyl)methacrylamide copolymer doxorubicin]: first member of a new class of chemotherapeutic agents-drug-polymer conjugates. Cancer Research Campaign Phase I/II Committee. Clinical cancer research : an official journal of the American Association for Cancer Research. 5 (1): 83-94 (1999).
[120] Seymour LW, Ferry DR, et al. Hepatic drug targeting: phase I evaluation of polymer-bound doxorubicin. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology. 20 (6): 1668-76 (2002).
[121] Gaillard PJ, Appeldoorn CCM, et al. Pharmacokinetics, Brain Delivery, and Efficacy in Brain Tumor-Bearing Mice of Glutathione Pegylated Liposomal Doxorubicin (2B3-101). PLoS One. 9 (1): e82331 (2014).
[122] Mamot C, Ritschard R, et al. Tolerability, safety, pharmacokinetics, and efficacy of doxorubicin-loaded anti-EGFR immunoliposomes in advanced solid tumours: a phase 1 dose-escalation study. The Lancet Oncology. 13 (12): 1234-41 (2012).
[123] McDonagh CF, Huhalov A, et al. Antitumor Activity of a Novel Bispecific Antibody That Targets the ErbB2/ErbB3 Oncogenic Unit and Inhibits Heregulin-Induced Activation of ErbB3. American Association for Cancer Research. 11 (3): 582-93 (2012).
[124] Matsumura Y, Hamaguchi T, et al. Phase I clinical trial and pharmacokinetic evaluation of NK911, a micelle-encapsulated doxorubicin. British Journal of Cancer. 91 (10): 1775-81 (2004).
[125] Valle JW, Armstrong A, et al. A phase 2 study of SP1049C, doxorubicin in P-glycoprotein-targeting pluronics, in patients with advanced adenocarcinoma of the esophagus and gastroesophageal junction. Investigational New Drugs. 29 (5): 1029-37 (2011).
[126] Barraud L, Merle P, et al. Increase of doxorubicin sensitivity by doxorubicin-loading into nanoparticles for hepatocellular carcinoma cells in vitro and in vivo. Journal of Hepatology. 42 (5): 736-43 (2005).
[127] Ho L-C, Hsu C-H, et al. Unibody core–shell smart polymer as a theranostic nanoparticle for drug delivery and MR imaging. Biomaterials. 37: 436-46 (2015).
[128] Jung ME, Lyster MA. Quantitative dealkylation of alkyl ethers via treatment with trimethylsilyl iodide. A new method for ether hydrolysis. The Journal of Organic Chemistry. 42 (23): 3761-4 (1977).
[129] Ben-Ishai D, Berger A. Cleavage of N-Carbobenzoxy Groups by dry Hydrogen Bromide and Hydrogen Chloride. The Journal of Organic Chemistry. 17 (12): 1564-70 (1952).
[130] Selvakannan PR, Swami A, et al. Synthesis of aqueous Au core-Ag shell nanoparticles using tyrosine as a pH-dependent reducing agent and assembling phase-transferred silver nanoparticles at the air-water interface. Langmuir. 20 (18): 7825-36 (2004).
[131] Si S, Bhattacharjee RR, et al. A mechanistic and kinetic study of the formation of metal nanoparticles by using synthetic tyrosine-based oligopeptides. Chemistry. 12 (4): 1256-65 (2006).
[132] Lehrer SS, Fasman GD. Ultraviolet irradiation effects in poly-L-tyrosine and model compounds. Identification of bityrosine as a photoproduct. Biochemistry. 6 (3): 757-67 (1967).
[133] Malencik DA, Sprouse JF, et al. Dityrosine: preparation, isolation, and analysis. Analytical Biochemistry. 242 (2): 202-13 (1996).
[134] Carlsen A, Lecommandoux S. Self-assembly of polypeptide-based block copolymer amphiphiles. Current Opinion in Colloid & Interface Science. 14 (5): 329-39 (2009).
[135] Lu Y, Mo R, et al. Self-folded redox/acid dual-responsive nanocarriers for anticancer drug delivery. Chemical Communications. 50 (95): 15105-8 (2014).
[136] Kataoka K, Harada A, et al. Block copolymer micelles for drug delivery: design, characterization and biological significance. Advanced Drug Delivery Reviews. 47 (1): 113-31 (2001).
[137] Davis ME, Chen Z, et al. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nature Reviews Drug Discovery. 7 (9): 771-82 (2008).
[138] Edmondson JM, Armstrong LS, et al. A rapid and simple MTT-based spectrophotometric assay for determining drug sensitivity in monolayer cultures. Journal of Tissue Culture Methods. 11 (1): 15-7 (1988).
[139] Ishiyama M, Shiga M, et al. A New Sulfonated Tetrazolium Salt That Produces a Highly Water-Soluble Formazan Dye. Chemical and Pharmaceutical Bulletin. 41 (6): 1118-22 (1993).
[140] Deng J, Gao N, et al. Self-assembled cationic micelles based on PEG-PLL-PLLeu hybrid polypeptides as highly effective gene vectors. Biomacromolecules. 13 (11): 3795-804 (2012).
[141] Wu L, Zou Y, et al. Intracellular release of doxorubicin from core-crosslinked polypeptide micelles triggered by both pH and reduction conditions. Biomaterials. 34 (21): 5262-72 (2013).
[142] Lv S, Tang Z, et al. Co-delivery of doxorubicin and paclitaxel by PEG-polypeptide nanovehicle for the treatment of non-small cell lung cancer. Biomaterials. 35 (23): 6118-29 (2014).
[143] Prabaharan M, Grailer JJ, et al. Amphiphilic multi-arm-block copolymer conjugated with doxorubicin via pH-sensitive hydrazone bond for tumor-targeted drug delivery. Biomaterials. 30 (29): 5757-66 (2009).
[144] Gillies ER, Fréchet JMJ. pH-Responsive Copolymer Assemblies for Controlled Release of Doxorubicin. Bioconjugate Chemistry. 16 (2): 361-8 (2005).
[145] Lv S, Song W, et al. Charge-conversional PEG-polypeptide polyionic complex nanoparticles from simple blending of a pair of oppositely charged block copolymers as an intelligent vehicle for efficient antitumor drug delivery. Molecular Pharmaceutics. 11 (5): 1562-74 (2014).
[146] Tomankova K, Polakova K, et al. In vitro cytotoxicity analysis of doxorubicin-loaded/superparamagnetic iron oxide colloidal nanoassemblies on MCF7 and NIH3T3 cell lines. International Journal of Nanomedicine. 10: 949-61 (2015).
[147] Lee GY, Song J-h, et al. Peptide-doxorubicin conjugates specifically degraded by matrix metalloproteinases expressed from tumor. Drug Development Research. 67 (5): 438-47 (2006).
[148] Hu F-Q, Wu X-l, et al. Cellular uptake and cytotoxicity of shell crosslinked stearic acid-grafted chitosan oligosaccharide micelles encapsulating doxorubicin. European Journal of Pharmaceutics and Biopharmaceutics. 69 (1): 117-25 (2008).
[149] Xiao W, Chen X, et al. Co-delivery of doxorubicin and plasmid by a novel FGFR-mediated cationic liposome. International Journal of Pharmaceutics. 393 (1–2): 120-7 (2010).
[150] Chen J-X, Wang M, et al. Hyaluronic acid and polyethylenimine self-assembled polyion complexes as pH-sensitive drug carrier for cancer therapy. Colloids and Surfaces B: Biointerfaces. 134: 81-7 (2015).
[151] Hirano K, Hunt CA. Lymphatic transport of liposome-encapsulated agents: effects of liposome size following intraperitoneal administration. Journal of Pharmaceutical Sciences. 74 (9): 915-21 (1985).
[152] Kohane DS, Tse JY, et al. Biodegradable polymeric microspheres and nanospheres for drug delivery in the peritoneum. Journal of Biomedical Materials Research Part A. 77 (2): 351-61 (2006).
[153] Hagiwara A, Takahashi T, et al. Clinical trials with intraperitoneal cisplatin microspheres for malignant ascites--a pilot study. Anti-cancer Drug Design. 8 (6): 463-70 (1993).
[154] Lukas G, Brindle SD, et al. The route of absorption of intraperitoneally administered compounds. The Journal of Pharmacology and Experimental Therapeutics. 178 (3): 562-4 (1971).
[155] Fang J, Sawa T, et al. Factors and mechanism of "EPR" effect and the enhanced antitumor effects of macromolecular drugs including SMANCS. Advances in Experimental Medicine and Biology. 519: 29-49 (2003).
[156] Fang J, Nakamura H, et al. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced Drug Delivery Reviews. 63 (3): 136-51 (2011).
[157] Maeda H, Fang J, et al. Vascular permeability enhancement in solid tumor: various factors, mechanisms involved and its implications. International Immunopharmacology. 3 (3): 319-28 (2003).
[158] Abuchowski A, McCoy JR, et al. Effect of covalent attachment of polyethylene glycol on immunogenicity and circulating life of bovine liver catalase. The Journal of Biological Chemistry. 252 (11): 3582-6 (1977).
[159] Fischer AH, Jacobson KA, et al. Hematoxylin and eosin staining of tissue and cell sections. Cold Spring Harbor protocols. 2008: pdb.prot4986 (2008).
[160] Zeiss CJ. The apoptosis-necrosis continuum: insights from genetically altered mice. Veterinary Pathology. 40 (5): 481-95 (2003).
[161] Elmore S. Apoptosis: A Review of Programmed Cell Death. Toxicologic Pathology. 35 (4): 495-516 (2007).
[162] Kerr JF, Wyllie AH, et al. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer. 26 (4): 239-57 (1972).
[163] Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. The American Journal of Pathology. 146 (1): 3-15 (1995).
[164] Trump BF, Berezesky IK, et al. The pathways of cell death: oncosis, apoptosis, and necrosis. Toxicologic Pathology. 25 (1): 82-8 (1997).
[165] Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature. 407 (6805): 784-8 (2000).
[166] Kurosaka K, Takahashi M, et al. Silent cleanup of very early apoptotic cells by macrophages. Journal of Immunology. 171 (9): 4672-9 (2003).
[167] Feoktistova M, Leverkus M. Programmed necrosis and necroptosis signalling. The FEBS Journal. 282 (1): 19-31 (2015).
[168] Havell EA, Fiers W, et al. The antitumor function of tumor necrosis factor (TNF), I. Therapeutic action of TNF against an established murine sarcoma is indirect, immunologically dependent, and limited by severe toxicity. The Journal of Experimental Medicine. 167 (3): 1067-85 (1988).
[169] Ho-Tin-Noé B, Carbo C, et al. Innate Immune Cells Induce Hemorrhage in Tumors during Thrombocytopenia. The American Journal of Pathology. 175 (4): 1699-708 (2009).
[170] Chen H, Li B, et al. Multifunctional near-infrared-emitting nano-conjugates based on gold clusters for tumor imaging and therapy. Biomaterials. 33 (33): 8461-76 (2012).
[171] Bulucu F, Ocal R, et al. Effects of N-Acetylcysteine, Deferoxamine and Selenium on Doxorubicin-Induced Hepatotoxicity. Biological Trace Element Research. 132 (1): 184-96 (2009).
[172] Singla DK, Ahmed A, et al. Embryonic stem cells improve cardiac function in Doxorubicin-induced cardiomyopathy mediated through multiple mechanisms. Cell Transplantation. 21 (9): 1919-30 (2012).
[173] Aldahmash BA, El-Nagar DM. Antioxidant effects of captopril against lead acetate-induced hepatic and splenic tissue toxicity in Swiss albino mice. Saudi Journal of Biological Sciences.
[174] Hira SK, Mondal I, et al. Downregulation of STAT3 phosphorylation enhances tumoricidal effect of IL-15-activated dendritic cell against doxorubicin-resistant lymphoma and leukemia via TNF-α. The International Journal of Biochemistry & Cell Biology. 67: 1-13 (2015).
[175] Chmielewska M, Symonowicz K, et al. Expression of metallothioneins I and II in kidney of doxorubicin-treated rats. Experimental & Toxicologic Pathology. 67 (4): 297-303 (2015).