簡易檢索 / 詳目顯示

研究生: 嚴家慶
Yen, Chia-Ching
論文名稱: 增加腦血管通透性以促進腦腫瘤治療之研究
Increasing Cerebrovascular Permeability to Facilitate Brain Tumor Therapy
指導教授: 蔡坤哲
Tsai, Kuen-Jer
謝清河
Hsieh, C.H. Patrick
學位類別: 碩士
Master
系所名稱: 醫學院 - 臨床醫學研究所
Institute of Clinical Medicine
論文出版年: 2014
畢業學年度: 102
語文別: 英文
論文頁數: 39
中文關鍵詞: 多型性神經膠母細胞瘤血腦障壁通透性
外文關鍵詞: glioblastoma multiforme, blood brain barrier, permeability
相關次數: 點閱:117下載:0
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
  • 多型性神經膠母細胞瘤是一種臨床上最常見且生長速度很快的惡性腦瘤,它也是一種具高度組織侵略性和浸潤性的致命癌症。此外,多型性神經膠母細胞瘤的病人平均存活率只有12到15個月,其生活機能狀態也較差。目前多型性神經膠母細胞瘤標準的治療方式是以手術、放射線治療與化療為主。然而,即使病人接受了標準的治療療程,其兩年平均存活率也僅上升了10%。另一方面,藥物治療也遇到了許多挑戰。而當腫瘤擴散到其他腦組織時,手術的方式將無法徹底的清除腫瘤。此外,血腦障壁的存在則是另一個挑戰,它防止且限制了許多藥物進入到腦組織,從而影響到藥物在腫瘤的傳遞與作用。目前儘管許多抗癌藥物像是帝盟多(Temozolomide)與癌思停(Bevacizumab)較能穿過血腦障壁,這些藥物也造成了許多副作用,像是高血壓、疲勞與腹瀉。
    在我的論文研究中,我們提供了一個新的思考方向--藉由血管生長因子(VEGF-A165)的前處理去改善抗癌藥物在血腦障壁的通透性與治療效果。我們的實驗結果證明了預先給予血管生長因子可以幫助藥物通過血腦障壁,進而增加了抗癌藥物的傳遞並降低了副作用。在伊文思藍(Evans blue)與奈米粒子的系統中,我們也證明了預先給予血管生長因子可以暫時性的增加血腦障壁的通透性,進而增加了伊文思藍與奈米粒子進入到腦組織中的比率。此外,我們也證明了預先給予血管生長因子再給予藥物跟單純只給予藥物的組別相比,可以明顯減少腫瘤體積、並且延長腫瘤老鼠的存活率。綜合以上結果,我們相信這項研究結果提供了未來腦瘤治療的新方向。

    Glioblastoma Multiforme (GBM) is the most common and the most fast-growing type of malignant brain tumor in clinical. It is a fatal cancer with high invasion and infiltration of the surrounding normal tissue. In addition, the GBM patients’ average survival rate is only 12-15 months and they also show poor functional health status. The current standard treatment is surgical resection, radiotherapy and chemotherapy. However, even if the patients receive these standard treatments, this median survival for adults only increases by about 10%, increasing the average survival rate to two years. Medical treatments for GBM also face several challenges. As the tumor is spread throughout the brain tissue, surgical resection is not able to completely remove it. In addition, the blood brain barrier (BBB) presents another challenge, preventing or limiting many chemotherapeutics from entering the brain tissue, thus affecting the drug delivery and effect in the tumor. Currently, new anti-cancer drugs such as Temozolomide (TMZ) or Bevacizumab (Avastin) have been developed which are able to penetrate the BBB more easily. However, these drugs still have some side effects, such as hypertension, fatigue and diarrhea.
    In this study, we provide a new direction to improve anticancer drug permeability through the BBB and enhance the therapeutic effects by pre-treatment with vascular endothelial growth factor-A165 (VEGF-A165). The results demonstrate that pre-treatment with VEGF-A165 could help drugs to penetrate the BBB and thus enhance drug delivery and reduce side effects. Using Evans blue and a nanoparticle-based system, we also show that pre-treatment with VEGF-A165 can temporarily increase BBB permeability and thus the ratio of dye and nanoparticles penetrate into the brain. Furthermore, we also show that pre-treatment with VEGF-A165 followed by TMZ significantly reduces the tumor size and prolongs the mean survival rate of mice with glioblastoma, compared to TMZ alone. In summary, we believe that the results of this study provide a new direction for the future of brain tumor therapy.

    Abstract.........I 摘要..........III 誌謝..........IV Table of Contents........V Index of Figures.........VIII Index of Tables..........IX Chapter 1...........1 Introduction...........1 1.1 Brain tumor..........1 1.1.1 Glioblastoma Multiforme (GBM).....1 1.1.2 Treatment Strategies of GBM......1 1.1.2.1 Surgical Resection and Radiation Therapy...1 1.1.2.2 Chemotherapy Drug in GBM Treatment.....2 1.1.2.2.1 Doxorubicin (DOX).....2 1.1.2.2.2 Bevacizumab (Avastin)....3 1.1.2.2.3 Temozolomide (TMZ)......3 1.2 Blood-Brain Barrier (BBB).......4 1.2.1 The Structure and Function of BBB.....4 1.2.2 The Limitations of Anticancer Drugs by BBB.....5 1.3 Methods for Increasing BBB Permeability and Drugs Delivery..5 1.3.1 Focus-Ultrasound.........5 1.3.2 Nanocarrier........6 1.3.3 Factors-TNF-alpha, IL-1beta,VEGF-A165......6 Chapter 2...........8 Hypothesis ...........8 Chapter 3..........9 Materials and Methods.........9 3.1 Animal.........9 3.2 Determination of BBB Permeability by Evans Blue Assay..9 3.3 Evans Blue Extraction and Analysis.......9 3.4 Nanoparticles Preparation and Characterization......9 3.5 Animals and Nanoparticle Injection.......10 3.6 Cell Culture.........10 3.7 Luciferase Lentivirus Backbone Plasmid Construction.10 3.8 Cell Viability Assay........11 3.9 Orthotropic Xenograft Animal Model.....11 3.10 Serum GPT, GOT, BUN and CRE Levels, Hematological Assessment Affected by TMZ........11 3.11 The Effect of Treatment on Tumor Size and Survival Rate...12 3.12 Statistical Analysis.......12 Chapter 4..........13 Results...........13 4.1 Systemic Injection of VEGF Increases the Permeability of the BBB in Mice.........13 4.2 Development of PEGylated Nanoparticles for Biocompatibility.15 4.3 Pretreatment of VEGF Improves Different sized PEGylated Fluorescent Nanoparticle Biodistribution and Penetration through the BBB..19 4.4 The Establishment of GBM Xenograft Model.....22 4.5 VEGF Pretreatment Followed by TMZ Injection Effectively Delays GBM Tumor Growth and Prolong Survival Rate in a GBM Mouse Model..........26 Chapter 5..........30 Discussion...........30 Chapter 6.........32 Conclusion and Future Work......32 Chapter 7.........33 References...........33 Index of Figures Figure 1. Systemic Injection of VEGF Increases the Permeability of the BBB in Mice........14 Figure 2. Development of PEGylated Nanoparticles for Biocompatibility..16 Figure 3. The TEM Images of Different Sizes PEGylated Nanoparticles..17 Figure 4. Pretreatment of VEGF Improves Different Sized PEGylated Fluorescent Nanoparticle Biodistribution and Penetration through the BBB..........20 Figure 5. Immunofluorescent Staining of Pretreatment with VEGF Improves Different Sized PEGylated Fluorescent Nanoparticles Penetrate through the BBB.....21 Figure 6. U-87 Cells were Transfected with the Luciferase Construct.23 Figure 7. The Establishment of GBM Xenograft Model...24 Figure 8. VEGF Pretreatment Followed by TMZ Injection Effectively Delays GBM Tumor Growth and Prolong Survival Rate in a GBM Mouse Model..........27 Figure 9. The IVIS Images of VEGF Pretreatment Followed by TMZ Injection Effectively Reduce GBM Tumor Growth in a GBM Mouse Model..........29 Index of Tables Table 1. Dynamic Light Scattering Studies of PEG-modified PS Particles..18 Table2. Transmission Electron Microscopy Studies of PEG-modified PS Particles.........18 Table 3. Blood Biochemical Data of Mice Infected and Treated with TMZ (Temozolomide) and VEGF......25 Table 4. Serum Biochemical Data of Mice Infected and Treated with TMZ (Temozolomide) and VEGF.......25 Table 5. VEGF Pretreatment Improve GBM Mouse Survival....29

    1.David N. Louis et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114: pp. 97–109, 2007.
    2.Hai Yan, M.D. et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 360: pp. 765–773, 2009.
    3.Shideng Bao et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444: pp.756-760, 2006.
    4.D. Williams Parsons et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 321: pp. 1807-1812, 2008.
    5.Croitor Sava et al. Ex vivo high resolution magic angle spinning metabolic profiles describe intratumoral histopathological tissue properties in adult human gliomas. Magnetic Resonance in Medicine. 65: pp. 320-328, 2011.
    6.Yunge Zhao et al. An extensive invasive intracranial human glioblastoma xenograft model: role of high level matrix metalloproteinase 9. Am J Pathol. 176: pp. 3032-3049, 2010.
    7.Federica Verginelli et al. Transcription factors FOXG1 and Groucho/TLEpromote glioblastoma growth. Nat Commun.4: pp. 2956, 2013.
    8.Clovis O. da Fonseca et al. Efficacy of monoterpene perillyl alcohol upon survival rate of patients with recurrent glioblastoma. J Cancer Res Clin Oncol. 137: pp. 287-293, 2011.
    9.J. Polivka et al. Isocitrate Dehydrogenase-1 Mutations as Prognostic Biomarker in Glioblastoma Multiforme Patients in West Bohemia. BioMed Research International.2014, 2014
    10.R. Stupp et al. “Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomized phase III study: 5-year analysis of the EORTC-NCIC trial. The
    Lancet Oncology. 10: pp. 459–466, 2009.
    11.Alba A. Brandes et al. Disease progression or pseudoprogression after concomitant radiochemotherapy treatment: Pitfalls in neurooncology. Neuro Oncol. 10: pp.361-367, 2008.
    12.Preusser M et al. Current concepts and management of glioblastoma. Ann Neurol . 70: pp. 9–21, 2011.
    13.Clarke J et al. Recent advances in therapy for glioblastoma. Arch Neurol. 67: pp. 279–283, 2010.
    14.Stupp, R et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352: pp. 987–996, 2005.
    15.P Liu et al. Cytotoxic effect of disulfiram/copper on human glioblastoma cell lines and ALDH-positive cancer-stem-like cells. British Journal of Cancer. 107: pp, 1488–1497, 2012.
    16.Tracy T. Batchelor et al. Age-dependent prognostic effects of genetic alterations in glioblastoma. Clin Cancer Res. 10: pp. 228–233, 2004.
    17.Carmine Cerullo et al. Complete remission and long term survival in recurrent malignant glioblastoma treated with fotemustine monotherapy: a case report. J Canc Ther. 4: pp. 584-587, 2013.
    18.Eyüpoglu IY et al. Surgical resection of malignant gliomas-role in optimizing patient outcome. Nat Rev Neurol. 9: pp. 141-51, 2013.
    19.N. Laperriere et al. Radiotherapy for newly diagnosed malignant glioma in adults a systematic review. Radiother Oncol. 64: pp. 259–273, 2002.
    20.Rajamanickam Baskar et al. Cancer and radiation therapy: current advances and future directions. Int J Med Sci. 9: pp.193-199, 2012.
    21.A. Narayana et al. Intensity-modulated radiotherapy in high-grade gliomas: clinical and dosimetric results. Int J Radiat Oncol Biol Phys. 64: pp. 892–897, 2006.
    22.Medical Research Council Brain Tumor Working Party. Randomized trial of procarbazine, lomustine, and vincristine in the adjuvant treatment of high grade astrocytoma: a medical research council trial. J Clin Oncol.19: pp.509 –518, 2001.
    23.Jing-Yi Chen et al. Quercetin-induced cardioprotection against doxorubicin cytotoxicity. J Biomed Sci. 20: pp. 95, 2013.
    24.Eirik Hagtvet et al. Liposomal doxorubicin improves radiotherapy response in hypoxic prostate cancer xenografts. Radiat Oncol. 6: pp. 135, 2011.
    25.Gouazé V et al. Glutathione peroxidase-1 overexpression prevents ceramide production and partially inhibits apoptosis in doxorubicin-treated human breast carcinoma cells. Mol Pharmacol. 60: pp. 488-496, 2001.
    26.Zhang S et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 18: pp. 1639-1642, 2012.
    27.Kim SY et al. Doxorubicin-induced reactive oxygen species generation and intracellular Ca2+ increase are reciprocally modulated in rat cardiomyocytes. Exp Mol Med. 38: pp. 535-545, 2006.
    28.ML Pinzón-Daza et al. The association of statins plus LDL receptor-targeted liposome-encapsulated doxorubicin increases in vitro drug delivery across blood–brain barrier cells. Br J Pharmacol. 167: pp. 1431–1447, 2012.
    29.Monnaert V et al. Effects of gamma- and hydroxypropyl-gamma-cyclodextrins on the transport of doxorubicin across an in vitro model of blood-brain barrier. J Pharmacol Exp Ther. 311: pp. 1115-1120, 2004.
    30.Vredenburgh JJ et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 25: pp. 4722-4729, 2007.
    31.Cohen MH et al. FDA drug approval summary: bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme. Oncologist. 14: pp. 1131-1138, 2009.
    32.Mark R. Gilbert et al. A randomized trial of bevacizumab for newly Diagnosed glioblastoma. N Engl J Med. 370: pp. 699-708, 2014.
    33.Qi Zhang et al. Monoclonal antibodies as therapeutic agents in oncology and antibody gene therapy. Cell Res. 17: pp. 89-99, 2007.
    34.Yuan Su et al. Effect of Angiogenesis Inhibitor Bevacizumab on Survival in Patients with Cancer: A Meta-Analysis of the Published Literature. PLoS ONE. 7: pp. e35629, 2012.
    35.Pàez-Ribes M et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 15: pp. 220-231, 2009.
    36.James L Rubenstein et al. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2: pp. 306–314, 2000.
    37.Norden AD et al. Antiangiogenic therapy for high-grade gliomas. Nat Rev Neurol. 5: pp.610-620, 2009.
    38.Verbeek B et al. O6-Methylguanine-DNA methyltransferase inactivation and chemotherapy. Br Med Bull. 85: pp. 17-33, 2008.
    39.Wick W et al. New (alternative) temozolomide regimens for the treatment of glioma. Neuro Oncol. 11: pp.69-79, 2009.
    40.Ohka F et al. Current trends in targeted therapies for glioblastoma multiforme. Neurol Res Int. 2012: pp. 878425, 2012.
    41.Goellner EM et al.. Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD+ biosynthesis and base excision repair. Cancer Res. 71: pp. 2308-2317, 2011.
    42.Munoz JL et al. Temozolomide resistance in glioblastoma cells occurs partly through epidermal growth factor receptor-mediated induction of connexin 43. Cell Death Dis. 5: pp. e1145, 2014.
    43.Kuo-Chen Wei et al. Focused ultrasound-induced blood–brain barrier opening to Enhance temozolomide delivery for glioblastoma treatment: a preclinical study. PLoS ONE. 8: pp e58995, 2013.
    44.Dagmar Beier et al. Efficacy of clinically relevant temozolomide dosing schemes in glioblastoma cancer stem cell lines. J Neurooncol. 109: pp. 45–52, 2012.
    45.Hawkins RA et al. Structure of the blood-brain barrier and its role in the transport of amino acids. J Nutr. 136: pp. 218S-26S, 2006.
    46.Abbott NJ et al. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 7: pp. 41-53, 2006.
    47.Li JY et al. Blood-brain barrier genomics. J Cereb Blood Flow Metab. 21: pp.61-68, 2001.
    48.Wohlfart S et al. Transport of drugs across the blood–brain barrier by nanoparticles. J Control Release. 161: pp. 264-273, 2012.
    49.Pardridge WM. Drug transport across the blood–brain barrier. J Cereb Blood Flow Metab. 32: pp.1959-1972, 2012.
    50.Park DM et al. Disposition of temozolomide in a patient with glioblastoma multiforme after gastric bypass surgery. J Neurooncol. 93: pp. 279-283, 2009.
    51.Gaya A et al. The use of temozolomide in recurrent malignant gliomas. Cancer Treat Rev. 28: pp. 115-120, 2002.
    52.Hynynen K et al. Local and reversible blood-brain barrier disruption by noninvasive focused ultrasound at frequencies suitable for trans-skull sonications. Neuroimage. 24:pp. 12-20, 2005.
    53.Liu HL et al. Hemorrhage detection during focused-ultrasound induced blood-brain-barrier opening by using susceptibility-weighted magnetic resonance imaging. Ultrasound Med Biol. 34: pp. 598–606, 2008.
    54.McDannold N et al. Targeted disruption of the blood-brain barrier with focused ultrasound: association with cavitation activity. Phys Med Biol. 51: pp. 793-807.
    55.McDannold N et al. Temporary disruption of the blood-brain barrier by use of ultrasound and microbubbles: safety and efficacy evaluation in rhesus macaques. Cancer Res. 72: pp. 3652-3663, 2012.
    56.Liu HL et al. Combining microbubbles and ultrasound for drug delivery to brain tumors: current progress and overview. Theranostics. 4: pp. 432-444, 2014.
    57.Cho K et al. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res. 14: pp. 1310-1316, 2008.
    58.Faraji AH et al. Nanoparticles in cellular drug delivery. Bioorg Med Chem. 17: pp. 2950-2962, 2009.
    59.Choi HS et al. Nanoparticles for biomedical imaging: fundamentals of clinical translation. Mol Imaging. 9: pp. 291-310, 2010.
    60.Argaw AT et al. IL-1beta regulates blood-brain barrier permeability via reactivation of the hypoxia-angiogenesis program. J Immunol. 177: pp. 5574-5584, 2006.
    61.Banks WA et al. Regional transport of TNF-alpha across the blood-brain barrier in young ICR and young and aged SAMP8 mice. Neurobiol Aging. 22: pp. 671-676, 2001.
    62.Ay I et al. VEGF increases blood–brain barrier permeability to Evans blue dye and tetanus toxin fragment C but not adeno-associated virus in ALS mice. Brain Res. 1234: pp. 198-205, 2008.
    63.Matsuno H et al. The role of TNF-alpha in the pathogenesis of inflammation and joint destruction in rheumatoid arthritis (RA): a study using a human RA/SCID mouse chimera. Rheumatology (Oxford). 41: pp. 329-337, 2002.
    64.Kistowska M et al. IL-1β drives inflammatory responses to propionibacterium acnes in vitro and in vivo. J Invest Dermatol. 134: pp. 677-685, 2014.
    65.Ferrara N et al. The biology of VEGF and its receptors. Nat Med. 9: pp. 669-676, 2003.
    66.Sibson NR et al. TNF-alpha reduces cerebral blood volume and disrupts tissue homeostasis via an endothelin- and TNFR2-dependent pathway. Brain. 125: pp. 2446-2459, 2002.
    67.Cui W et al. Alterations of blood-brain barrier and associated factors in acute liver failure. Gastroenterol Res Pract. 2013: pp. 841707, 2013.
    68.Eming SA et al. Molecular mechanisms of VEGF-A action during tissue repair. J Investig Dermatol Symp Proc. 11: pp. 79-86, 2006.
    69.Valable S et al. VEGF-induced BBB permeability is associated with an MMP-9 activity increase in cerebral ischemia: both effects decreased by Ang-1. J Cereb Blood Flow Metab. 25: pp. 1491-1504, 2005.
    70.Lin YD et al. Instructive nanofiber scaffolds with VEGF create a microenvironment for arteriogenesis and cardiac repair. Sci Transl Med. 4: pp.146ra109, 2012.
    71.Zhao LN et al. Vascular endothelial growth factor increases permeability of the blood-tumor barrier via caveolae-mediated transcellular pathway. J Mol Neurosci. 44: pp. 122-129, 2011.
    72.Elizabeth A. Nance et al. A Dense Poly(Ethylene Glycol) Coating Improves Penetration of Large Polymeric Nanoparticles Within Brain Tissue. Sci Transl Med. 4: pp. 149ra119, 2012.
    73.Avraham-Davidi I et al. On-site education of VEGF-recruited monocytes improves their performance as angiogenic and arteriogenic accessory cells. J Exp Med. 210: pp. 2611-2625, 2013.
    74.Zentilin L et al. Bone marrow mononuclear cells are recruited to the sites of VEGF-induced neovascularization but are not incorporated into the newly formed vessels. Blood. 107: pp. 3546-3654, 2006.
    75.Redente EF et al. Tumor signaling to the bone marrow changes the phenotype of monocytes and pulmonary macrophages during urethane-induced primary lung tumorigenesis in A/J mice. Am J Pathol. 170: pp. 693-708, 2007.

    無法下載圖示 校內:2019-09-09公開
    校外:不公開
    電子論文尚未授權公開,紙本請查館藏目錄
    QR CODE