簡易檢索 / 詳目顯示

研究生: 莊子玄
Chuang, Tzu-Hsuan
論文名稱: 發展同時搭載鐵鉑奈米粒子和丙酮特安皮質醇的新型藥物傳遞系統並評估在蟹足腫上的協同治療效果
To develop a novel drug delivery system to carry both FePt nanoparticles and Triamcinolone acetonide and evaluate the synergistic effect of keloid treatment
指導教授: 吳炳慶
Wu, Ping-Ching
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: 2020
畢業學年度: 108
語文別: 英文
論文頁數: 79
中文關鍵詞: 蟹足腫細胞鐵鉑奈米粒子輻射療法輻射增敏劑聚乳酸-甘醇酸共聚物滲透和留滯作用丙酮特安皮質醇協同作用
外文關鍵詞: Keloid fibroblast(KF), iron-platinum nanoparticles (FePt NPs), radiation therapy, radiation sensitizer, poly(lactic-co-glycolic acid)PLGA, permeability and retention (EPR) effect, triamcinolone acetonide(TAC), synergistic-function
相關次數: 點閱:71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
  • 蟹足腫是一種由於傷口癒合不正常而引起的纖維化增生。其疤痕會生長超過原始傷口的範圍,不斷生長,很少隨時間消退,並且其在有色個體中最常見。雖然它是良性的,沒有惡性潛能,但是蟹足腫造成的外觀毀損會給病人相當大的身心不利影響。當前,仍然沒有能夠立即、完全和永久去除蟹足腫並恢復皮膚功能的治療方法。臨床上存在多種治療蟹足腫的方法,但這些方法都有其自身的優劣勢;然而,副作用和經常復發的風險仍然很高。因此,現今研究主要專注於改善原有的蟹足腫的治療方法或尋找新藥物。
    近年來,奈米材料的發展已大大改善了臨床應用中的診斷和治療。鐵鉑奈米粒子(FePt NPs)已被開發用於生物醫學應用中,因為它具有超順磁性,抗氧化性和極高化學穩定性等優異特性。而鐵鉑奈米粒子的高原子序數(Z)金屬性能讓它在X射線的放射線照射下,可作為臨床具有潛能的輻射增敏劑。另外,聚乳酸-甘醇酸共聚物(PLGA)對人體具有非常低的毒性,可以應用於生物醫學中的奈米藥物傳輸載體。而其奈米尺寸的聚乳酸-甘醇酸共聚物作為載體可以通過高強度的滲透和留滯作用(EPR),迅速在組織內擴散和積累。具體而言,我們打算借助奈米的藥物遞送功能,徹底改變蟹足腫是治療策略。
    這項研究的目的是利用聚乳酸-甘醇酸共聚物(PLGA)做為奈米藥物載體同時搭載第一線臨床類固醇藥物丙酮特安皮質醇(TAC)和輻射增敏劑鐵鉑金屬粒子,並測試其在蟹足腫上的治療效果。我們認為輻射增敏的鐵鉑奈米粒子可以增強放射治療的功效,而皮質類固醇,丙酮特安皮質醇,可以抑制蟹足腫細胞的發炎環境。因此,FePt-PLGA-TAC NPs將有機會是減少藥物劑量和並發症發生率的理想策略。
    在這項研究中,我們首先證明了在顯微鏡下,FePt-PLGA-TAC NPs形成了均勻而圓形的顆粒。粒子的大小為219.7±50.8 nm,表面電位為-24.38 ± 0.85 mV。TAC的包覆效率為63.75 ± 0.50 %,高於先前的PLGA-TAC NPs。而在蟹足腫細胞中的測試中,
    FePt-PLGA-TAC NPs的放射增敏作用,使其中細胞存活率在72 h時降至10 %,並顯著喪失了增殖能力。我們還觀察到,我們的FePt-PLGA-TAC NPs結合低輻射劑量,具有通過FePt NPs和TAC藥物之間的協同功能調節炎症反應的良好能力。重要的是,在奈米藥物的處理下,作為蟹足腫細胞中的I型膠原蛋白標誌其表現量也明顯下降。由於目前缺乏合適的動物模型,我們尚不能在動物層級上驗證FePt-PLGA-TAC NPs在蟹足腫治療上的功效。然而,從上述的細胞實驗結果,FePt-PLGA-TAC NPs將來依舊有潛力作為蟹足腫治療的藥物。

    Keloids are the fibroproliferative disorders of the skin that result from an abnormal wound healing response. Keloids can grow beyond the original borders of the wound, continue to grow, rarely regress, and are the most common in pigmented individuals. Although benign and with no malignant potential, disfigurement from keloids leads to considerable physical and psychological adverse effects. Currently, there are still no specific therapeutics that can instantly, completely, and permanently remove keloids and restore the function of the skin. A wide range of therapeutic options exist for the treatment of keloids, all of which have their strengths; however, a high risk of side-effects and frequent recurrence remains. Therefore, the present study aimed to identify improved therapeutic approaches to drugs for the treatment of keloids.
    In recent years, the development of nanomaterials has substantially improved diagnosis and treatment in clinical applications. Iron-platinum nanoparticles (FePt NPs) have been developed in biomedical applications due to their superior characteristics, including super-paramagnetism, resistance to oxidation, and high chemical stability. The high atomic number (Z) metallic property of FePt NPs under X-ray based radiotherapy beams can be a potential radiation sensitizer in clinical applications. Additionally, poly(lactic-co-glycolic acid) (PLGA) as a nanocarrier is considered to produce minimal systemic toxicity when used for biomedical applications. Owing to the nano-size, PLGA NPs as a carrier can rapidly spread and accumulate within tissues through the enhanced permeability and retention (EPR) effect. Specifically, we intend to revolutionize the strategy of keloid treatment through the use of advance of the nano-based drug delivery features.
    The purpose of this study is to develop a synergistic-functional FePt polymer nanoparticle with first-line steroid drug Triamcinolone acetonide (TAC) with the poly lactic-co-glycolic acid (PLGA). We assume that radiation-sensitive FePt NPs can enhance the efficacy of radiation therapy and that corticosteroid TAC can inhibit the inflammatory environment of keloid fibroblasts(KF). Thus, FePt-PLGA-TAC NPs might be an ideal strategy to reduce the prescribed dose and the incidence of complications.
    In this study, we first demonstrated that FePt-PLGA-TAC NPs formed uniform and round particles under the microscope. The particles were 219.7 ± 50.8 nm in size, and the zeta potential was -24.38 ± 0.85 mV. The encapsulation efficiency of TAC was 63.75 ± 0.50 %, which is greater than that of other common PLGA NPs. A test in KF cells using FePt-PLGA-TAC NPs enhanced the radio-sensitizing effects on KF, whose of cell viability was lowered to 10 % at 72 h, and dramatically decreased the proliferative ability. We also observed that our FePt-PLGA-TAC NPs combined with low radiation dosage had better ability to regulate the inflammatory reaction due to the synergistic function between FePt NPs and TAC drugs. Importantly, the hallmark of collagen type I expression in keloids was also expressed lower under the NPs treatment. Due to the absence of a suitable animal model, we still cannot prove the efficacy of FePt-PLGA-TAC NPs in keloid therapy. However, from these cell studies, we expect that FePt-PLGA-TAC NPs will be a promising agent for keloid treatment in the future.

    Content 摘要 I Abstract III 致謝 V Content VII List of tables X List of figures X Chapter 1 Introduction 1 1. The current problem of keloid treatment 1 1.1 Definition of Keloid Scar 1 1.2 Normal wound healing and scar formation 1 1.3 The pathogenesis of keloid 2 1.3.1 The TGF-β ⁄Smad signaling pathway 2 1.3.2 The IL-6 signaling pathway 3 1.3.3 The NF-κß signaling pathway 3 1.3.4 The Extracellular matrix(ECM) 4 1.4 The Problem of Clinical Treatment 4 1.4.1 Steroid therapy in keloid disease 5 1.4.2 Radiation therapy for keloid 5 1.5 The main limitation of therapeutic development of Keloid 6 1.6 Radiation therapy 7 1.6.1 Radiosensitizers 7 1.6.2 FePt nanoparticles 8 1.7 Polymeric nanoparticles delivery system 8 1.7.1 Drug delivery system 8 1.7.2 PLGA nanoparticle 9 1.8 Specific aims 9 Chapter 2 Materials and Methods 11 2.1 Material 11 2.2 Preparation of 12 nm FePt nanoparticles 12 2.3 Ligand Exchange of FePt nanoparticles 12 2.4 Preparation of the TAC loaded into PLGA nanoparticles 12 2.5 Determination of the content of TAC in PLGA nanoparticles 13 2.6 TAC release from PLGA nanoparticles 13 2.7 Preparation of the FePt-conjugated PLGA containing TAC 14 2.8 The physicochemical properties of the nanoparticles 14 2.8.1 The morphology and particle size of the nanoparticles 14 2.8.2 Zeta potential and Fourier transform infrared (FT-IR) spectrometry 15 2.9 Cell Culture 15 2.9.1 Fibroblasts Primary Cell Culture 15 2.9.2 The Cell Count 16 2.10 Preparation of Rhodamine B-Loaded Nanoparticles 16 2.11 Cellular uptake of RhB-loaded PLGA NPs 17 2.12 Cell viability assay 17 2.13 Electronic beam irradiation 17 2.14 Immunocytochemistry to detect Ki67 in the KF 18 2.15 Total ROS measurement in KF 18 2.16 Immunocytochemistry to detect nuclear factor kappa B (NF-κB) activation in KF… 19 2.17 Quantitative Determination of the mRNA expression in Collagen I of KF 20 2.18 Keloid explants are embedded in the collagen matrix 20 2.19 Apoptosis and Ki-67 Detection on Ex-vivo model by Immunohistochemistry.. 21 2.20 Data Analysis and Statistical Evaluation 21 Chapter 3 Results 22 3.1 Synthesis and characterization of the FePt-PLGA-TAC NPs 22 3.1.1 Synthesis and characterization of the FePt NPs 22 3.1.2 The size and shape of the distributions 22 3.1.3 The zeta potential and the FTIR spectra 22 3.2 Determination of TAC concentrations and drug release in FePt-PLGA-TAC NPs… 23 3.3 KF demonstrated cancel-like characteristic and resistance to radiotherapy compared to NF 24 3.4 The cytotoxicity of PLGA-TAC NPs 24 3.5 FePt-PLGA-TAC NPs enhanced the radiation sensitivity in KF 25 3.6 FePt-PLGA-TAC NPs influence the proliferation and cell number of KF 26 3.7 FePt-PLGA-TAC NPs regulate ROS in KF 27 3.8 FePt-PLGA-TAC NPs regulate inflammation in the KF 27 3.9 FePt-PLGA-TAC NPs had negative effects on collagen type I expression in KFs… 28 3.10 Ex vivo organ culturing of Keloid 28 Chapter 4 Discussion 30 4.1 Characterization of the FePt-PLGA-TAC NPs 30 4.2 The Cancer-like properties and radiation resistance in KF 31 4.3 FePt-PLGA-TAC NPs enhanced the radiation sensitivity in KFs 32 4.4 FePt-PLGA-TAC NPs regulate the ROS and inflammation in KF. 33 4.5 FePt-PLGA-TAC NPs reduce collagen production. 34 4.6 The challenge of achieving long-term culture of Keloid organ models 34 Chapter 5 Conclusion 36 Reference 72 List of tables Table 1. Characteristics of keloids scars[3-6]. 39 Table 2. Characterization of nanoparticle 40 Table 3: Characteristics of the individuals used in this study. 41 List of figures Figure 1. Typical keloid scars. 42 Figure 2. The process of normal wound healing. 42 Figure 3.Scheme for keloid pathogenesis and possible mechanisms. 43 Figure 4. Schematic illustration of the interaction between nanoparticles and ionizing radiation. ROS: reactive oxygen species[55]. 43 Figure 5: Schematic illustration of mechanism of synergistic-functional FePt-PLGA-TAC NPs for keloid treatment. 44 Figure 6. Polyol reduction method for the synthesis of FePt-Cys NPs. 44 Figure 7: Emulsion-evaporation method for synthesis of PLGA-TAC NPs. 45 Figure 8. The way to synthesize synergistic-functional FePt-PLGA-TAC NPs. 45 Figure 9. A flow chart for the preparation of primary cell cultures. 46 Figure 10: Schematic representation of the keloid explant culture. 47 Figure 11. The morphology and solubility test of FePt NPs. 48 Figure 12. TEM photomicrograph of (A) empty PLGA nanoparticles (B) PLGA-TAC nanoparticles (C) unmodified FePt nanoparticles (D)FePt-PLGA-TAC nanoparticles. 49 Figure 13. Particle size distribution of FePt-PLGA-TAC NPs was measured by DLS. 50 Figure 14. The surface charge of FePt-PLGA-TAC NPs was determined by zeta potential. 51 Figure 15: FTIR transmittance spectrum of (A) FePt-PLGA-TAC NPs (B) FePt-Cys NPs and (C) empty PLGA only NPs, full wave number from 400-4000 cm-1. 51 Figure 16. The Encapsulating rate of TAC in PLGA-TAC NPs. 52 Figure 17. In vitro drug release profiles of TAC from PLGA-TAC NPs. 53 Figure 18. Uptake kinetics of RhB-loaded PLGA NPs in NskinF and KF. 55 Figure 19. The resistance of ionizing radiation to NskinF、sNskinF and KF. 56 Figure 20. The cytotoxicity of PLGA-TAC NPs and TAC alone in NskinF、sNskinF and KF. 57 Figure 21. The cytotoxicity of FePt-PLGA NPs under radiation in sNskinF and KF. 58 Figure 22: FePt-PLGA-TAC NPs enhanced radio sensitization of KF. 59 Figure 23: Effects of FePt-PLGA-TAC NPs on KF with several doses of irradiation in reducing cell viability over time. 60 Figure 24 Effects of FePt-PLGA-TAC NPs with radiation on viable cell numbers at direct cell counting in KF over time. 61 Figure 25. Effects of FePt-PLGA-TAC NPs with radiation on Ki-67 immunofluorescence staining in KF cells. 62 Figure 26. The quantification of ROS generation over time. 64 Figure 27. Effects of FePt-PLGA-TAC NPs with radiation on inhibition of NF-κB activation in KF cells. 65 Figure 28. Immunofluorescence analysis of collagen type I at Day 7 post FePt-PLGA-TAC NPs with radiation treatment. 67 Figure 29.Quantitative real-time PCR measurments of collagen I mRNA expression levels at 72 h post treatment with FePt-PLGA-TAC NPs with radiation treatment group 69 Figure 30: Cell apoptosis using the TUNNEL assay of the Keloid ex-vivo model 70 Figure 31: Cell proliferation using the Ki-67 marker of Keloid in an ex-vivo model 70 Figure 32. Risk factors of keloid and the mechanism of the synergistic-functional FePt-PLGA-TAC NPs for keloid treatment. 71

    Reference
    1. Butler, P.D., M.T. Longaker, and G.P. Yang, Current progress in keloid research and treatment. Journal of the American College of Surgeons, 2008. 206(4): p. 731-741.
    2. Al-Attar, A., et al., Keloid Pathogenesis and Treatment. Plastic and Reconstructive Surgery, 2006. 117(1): p. 286-300.
    3. Gauglitz, G.G., et al., Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Mol Med, 2011. 17(1-2): p. 113-25.
    4. Shaheen, A., Comprehensive Review of Keloid Formation. Journal of Clinical Research in Dermatology, 2017. 4(5): p. 1-18.
    5. Gauglitz, G.G., et al., Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Molecular medicine, 2011. 17(1-2): p. 113-125.
    6. Sun, L.-M., K.-H. Wang, and Y.-C.G. Lee, Keloid incidence in Asian people and its comorbidity with other fibrosis-related diseases: a nationwide population-based study. Archives of dermatological research, 2014. 306(9): p. 803-808.
    7. Eming, S.A., P. Martin, and M. Tomic-Canic, Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med, 2014. 6(265): p. 265sr6.
    8. Xue, M. and C.J. Jackson, Extracellular Matrix Reorganization During Wound Healing and Its Impact on Abnormal Scarring. Adv Wound Care (New Rochelle), 2015. 4(3): p. 119-136.
    9. Gonzalez, A.C., et al., Wound healing - A literature review. An Bras Dermatol, 2016. 91(5): p. 614-620.
    10. Ogawa, R., Keloid and Hypertrophic Scars Are the Result of Chronic Inflammation in the Reticular Dermis. Int J Mol Sci, 2017. 18(3).
    11. Tuan, T.-L. and L.S. Nichter, The molecular basis of keloid and hypertrophic scar formation. Molecular medicine today, 1998. 4(1): p. 19-24.
    12. Shah, M., D.M. Foreman, and M. Ferguson, Neutralisation of TGF-beta 1 and TGF-beta 2 or exogenous addition of TGF-beta 3 to cutaneous rat wounds reduces scarring. Journal of cell science, 1995. 108(3): p. 985-1002.
    13. He, S., et al., Mechanisms of transforming growth factor beta(1)/Smad signalling mediated by mitogen-activated protein kinase pathways in keloid fibroblasts. Br J Dermatol, 2010. 162(3): p. 538-46.
    14. Brown, J.J. and A. Bayat, Genetic susceptibility to raised dermal scarring. Br J Dermatol, 2009. 161(1): p. 8-18.
    15. Tao, S. and K. Sampath, Alternative splicing of SMADs in differentiation and tissue homeostasis. Development, growth & differentiation, 2010. 52(4): p. 335-342.
    16. Ghazizadeh, M., Essential role of IL-6 signaling pathway in keloid pathogenesis. Journal of Nippon Medical School, 2007. 74(1): p. 11-22.
    17. Ghazizadeh, M., et al., Functional implications of the IL-6 signaling pathway in keloid pathogenesis. Journal of investigative Dermatology, 2007. 127(1): p. 98-105.
    18. Quong, W.L., Y. Kozai, and R. Ogawa, A case of keloids complicated by Castleman’s disease: interleukin-6 as a keloid risk factor. Plastic and Reconstructive Surgery Global Open, 2017. 5(5).
    19. O’Reilly, S., et al., Interleukin-6, its role in fibrosing conditions. Cytokine & growth factor reviews, 2012. 23(3): p. 99-107.
    20. Lim, C., et al., Stat3 contributes to keloid pathogenesis via promoting collagen production, cell proliferation and migration. Oncogene, 2006. 25(39): p. 5416-5425.
    21. Liu, T., et al., NF-κB signaling in inflammation. Signal transduction and targeted therapy, 2017. 2(1): p. 1-9.
    22. Mitchell, J.P. and R.J. Carmody, NF-κB and the transcriptional control of inflammation, in International review of cell and molecular biology. 2018, Elsevier. p. 41-84.
    23. Messadi, D.V., et al., Activation of NFκB signal pathways in keloid fibroblasts. Archives of dermatological research, 2004. 296(3): p. 125-133.
    24. Verhaegen, P.D., et al., Differences in collagen architecture between keloid, hypertrophic scar, normotrophic scar, and normal skin: an objective histopathological analysis. Wound Repair and Regeneration, 2009. 17(5): p. 649-656.
    25. Rohani, M.G. and W.C. Parks, Matrix remodeling by MMPs during wound repair. Matrix Biology, 2015. 44: p. 113-121.
    26. Ulrich, D., et al., Matrix metalloproteinases and tissue inhibitors of metalloproteinases in patients with different types of scars and keloids. Journal of Plastic, Reconstructive & Aesthetic Surgery, 2010. 63(6): p. 1015-1021.
    27. Roomi, M., et al., Patterns of MMP-2 and MMP-9 expression in human cancer cell lines. Oncology reports, 2009. 21(5): p. 1323-1333.
    28. Andrews, J.P., et al., Keloids: The paradigm of skin fibrosis—Pathomechanisms and treatment. Matrix Biology, 2016. 51: p. 37-46.
    29. Funayama, E., et al., Keratinocytes promote proliferation and inhibit apoptosis of the underlying fibroblasts: an important role in the pathogenesis of keloid. Journal of investigative dermatology, 2003. 121(6): p. 1326-1331.
    30. Hahn, J.M., et al., Keloid‐derived keratinocytes exhibit an abnormal gene expression profile consistent with a distinct causal role in keloid pathology. Wound Repair and Regeneration, 2013. 21(4): p. 530-544.
    31. Lim, I.J., et al., Investigation of the influence of keloid-derived keratinocytes on fibroblast growth and proliferation in vitro. Plastic and reconstructive surgery, 2001. 107(3): p. 797-808.
    32. Ogawa, R., et al., Keloids and hypertrophic scars can now be cured completely: recent progress in our understanding of the pathogenesis of keloids and hypertrophic scars and the most promising current therapeutic strategy. Journal of Nippon Medical School, 2016. 83(2): p. 46-53.
    33. Chung, K. and I. Adcock, Signalling and transcriptional regulation in inflammatory and immune cells: importance in lung biology and disease. 2005, Eur Respiratory Soc.
    34. Wong, T.-S., et al., The efficacy of triamcinolone acetonide in keloid treatment: a systematic review and meta-analysis. Frontiers in medicine, 2016. 3: p. 71.
    35. Coppola, M.M., et al., Triamcinolone acetonide intralesional injection for the treatment of keloid scars: patient selection and perspectives. Clinical, cosmetic and investigational dermatology, 2018. 11: p. 387.
    36. Roques, C. and L. Téot, The use of corticosteroids to treat keloids: a review. The international journal of lower extremity wounds, 2008. 7(3): p. 137-145.
    37. Muneuchi, G., et al., Long-term outcome of intralesional injection of triamcinolone acetonide for the treatment of keloid scars in Asian patients. Scandinavian journal of plastic and reconstructive surgery and hand surgery, 2006. 40(2): p. 111-116.
    38. Arno, A.I., et al., Up-to-date approach to manage keloids and hypertrophic scars: a useful guide. Burns, 2014. 40(7): p. 1255-1266.
    39. Ogawa, R., Keloids and Hypertrophic Scars Can Now Be Treated Completely by Multimodal Therapy, Including Surgery, Followed by Radiation and Corticosteroid Tape/Plaster, in Scars. 2019, IntechOpen.
    40. Huang, C., et al., Managing keloid scars: From radiation therapy to actual and potential drug deliveries. International wound journal, 2019. 16(3): p. 852-859.
    41. Jones, M.E., C. Hardy, and J. Ridgway, Keloid management: a retrospective case review on a new approach using surgical excision, platelet-rich plasma, and in-office superficial photon X-ray radiation therapy. Advances in skin & wound care, 2016. 29(7): p. 303.
    42. Sruthi, K., et al., Single-fraction radiation: A promising adjuvant therapy to prevent keloid recurrence. Journal of cancer research and therapeutics, 2018. 14(6): p. 1251.
    43. Sharma, J.R., et al., In Vitro and Ex Vivo Models for Functional Testing of Therapeutic Anti-scarring Drug Targets in Keloids. Advances in Wound Care, 2019. 8(12): p. 655-670.
    44. van den Broek, L.J., et al., Human hypertrophic and keloid scar models: principles, limitations and future challenges from a tissue engineering perspective. Experimental dermatology, 2014. 23(6): p. 382-386.
    45. Tsai, C.-H. and R. Ogawa, Keloid research: current status and future directions. Scars, Burns & Healing, 2019. 5: p. 2059513119868659.
    46. Syed, F. and A. Bayat, Superior effect of combination vs. single steroid therapy in keloid disease: a comparative in vitro analysis of glucocorticoids. Wound Repair and Regeneration, 2013. 21(1): p. 88-102.
    47. Bagabir, R., et al., Long‐term organ culture of keloid disease tissue. Experimental dermatology, 2012. 21(5): p. 376-381.
    48. Citrin, D.E., Recent Developments in Radiotherapy. N Engl J Med, 2017. 377(11): p. 1065-1075.
    49. Stapleton, S., D. Jaffray, and M. Milosevic, Radiation effects on the tumor microenvironment: Implications for nanomedicine delivery. Adv Drug Deliv Rev, 2017. 109: p. 119-130.
    50. Lee, M.H., et al., Disulfide-cleavage-triggered chemosensors and their biological applications. Chemical reviews, 2013. 113(7): p. 5071-5109.
    51. Jeremic, B., A.R. Aguerri, and N. Filipovic, Radiosensitization by gold nanoparticles. Clinical and Translational Oncology, 2013. 15(8): p. 593-601.
    52. Porcel, E., et al., Platinum nanoparticles: a promising material for future cancer therapy? Nanotechnology, 2010. 21(8): p. 085103.
    53. Mirjolet, C., et al., The radiosensitization effect of titanate nanotubes as a new tool in radiation therapy for glioblastoma: a proof-of-concept. Radiotherapy and Oncology, 2013. 108(1): p. 136-142.
    54. Kobayashi, K., et al., Enhancement of radiation effect by heavy elements. Mutat Res, 2010. 704(1-3): p. 123-31.
    55. Bergs, J.W., et al., The role of recent nanotechnology in enhancing the efficacy of radiation therapy. Biochim Biophys Acta, 2015. 1856(1): p. 130-43.
    56. Xie, J., et al., Emerging Strategies of Nanomaterial-Mediated Tumor Radiosensitization. Adv Mater, 2019. 31(3): p. e1802244.
    57. Zheng, Y., et al., FePt nanoparticles as a potential X-ray activated chemotherapy agent for HeLa cells. Int J Nanomedicine, 2015. 10: p. 6435-44.
    58. Shi, Y., et al., Recent Advances in FePt Nanoparticles for Biomedicine. Journal of Nanomaterials, 2015: p. 1-13.
    59. Rajendran, L., H.-J. Knölker, and K. Simons, Subcellular targeting strategies for drug design and delivery. Nature reviews Drug discovery, 2010. 9(1): p. 29-42.
    60. Ferrari, R., et al., Polymer nanoparticles for the intravenous delivery of anticancer drugs: the checkpoints on the road from the synthesis to clinical translation. Nanoscale, 2018. 10(48): p. 22701-22719.
    61. Acharya, S. and S.K. Sahoo, PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect. Advanced drug delivery reviews, 2011. 63(3): p. 170-183.
    62. Paulo, C.S., R.P. das Neves, and L.S. Ferreira, Nanoparticles for intracellular-targeted drug delivery. Nanotechnology, 2011. 22(49): p. 494002.
    63. El-Sawy, H.S., et al., Stimuli-responsive nano-architecture drug-delivery systems to solid tumor micromilieu: past, present, and future perspectives. ACS nano, 2018. 12(11): p. 10636-10664.
    64. Gao, Y., et al., Transdermal delivery of 10, 11-methylenedioxycamptothecin by hyaluronic acid based nanoemulsion for inhibition of keloid fibroblast. Carbohydrate polymers, 2014. 112: p. 376-386.
    65. Elmowafy, E.M., M. Tiboni, and M.E. Soliman, Biocompatibility, biodegradation and biomedical applications of poly (lactic acid)/poly (lactic-co-glycolic acid) micro and nanoparticles. Journal of Pharmaceutical Investigation, 2019: p. 1-34.
    66. Yen, C.-L., et al., Targeted Delivery of Curcumin Rescues Endoplasmic Reticulum–Retained Mutant NOX2 Protein and Avoids Leukocyte Apoptosis. The Journal of Immunology, 2019. 202(12): p. 3394-3403.
    67. Mundargi, R.C., et al., Nano/micro technologies for delivering macromolecular therapeutics using poly (D, L-lactide-co-glycolide) and its derivatives. Journal of Controlled Release, 2008. 125(3): p. 193-209.
    68. Hernández-Giottonini, K.Y., et al., PLGA nanoparticle preparations by emulsification and nanoprecipitation techniques: effects of formulation parameters. RSC Advances, 2020. 10(8): p. 4218-4231.
    69. Pereira, A., et al., Development and validation of a high performance liquid chromatographic method for determination of triamcinolone acetonide from polyurethane intraocular implants. Int. J. Pharm. Pharm. Sci, 2012. 4(4): p. 132-136.
    70. Matysova, L., et al., Determination of methylparaben, propylparaben, triamcinolone acetonide and its degradation product in a topical cream by RP-HPLC. Analytical and bioanalytical chemistry, 2003. 376(4): p. 440-443.
    71. Kraisit, P. and N. Sarisuta, Development of Triamcinolone Acetonide-Loaded Nanostructured Lipid Carriers (NLCs) for Buccal Drug Delivery Using the Box-Behnken Design. Molecules, 2018. 23(4): p. 982.
    72. Tamboli, V., G.P. Mishra, and A.K. Mitra, Novel pentablock copolymer (PLA–PCL–PEG–PCL–PLA)-based nanoparticles for controlled drug delivery: effect of copolymer compositions on the crystallinity of copolymers and in vitro drug release profile from nanoparticles. Colloid and polymer science, 2013. 291(5): p. 1235-1245.
    73. Hsu, C.-K., et al., Caveolin-1 controls hyperresponsiveness to mechanical stimuli and fibrogenesis-associated RUNX2 activation in keloid fibroblasts. Journal of Investigative Dermatology, 2018. 138(1): p. 208-218.
    74. Jonderian, A. and R. Maalouf, Formulation and in vitro interaction of rhodamine-B loaded PLGA nanoparticles with cardiac myocytes. Frontiers in pharmacology, 2016. 7: p. 458.
    75. Blechinger, J., et al., Uptake kinetics and nanotoxicity of silica nanoparticles are cell type dependent. Small, 2013. 9(23): p. 3970-3980.
    76. Sun, Y., et al., FePt-Cys nanoparticles induce ROS-dependent cell toxicity, and enhance chemo-radiation sensitivity of NSCLC cells in vivo and in vitro. Cancer letters, 2018. 418: p. 27-40.
    77. Noursadeghi, M., et al., Quantitative imaging assay for NF-κB nuclear translocation in primary human macrophages. Journal of immunological methods, 2008. 329(1-2): p. 194-200.
    78. Fejes, Z., et al., Endothelial cell activation is attenuated by everolimus via transcriptional and post-transcriptional regulatory mechanisms after drug-eluting coronary stenting. PloS one, 2018. 13(6).
    79. Syed, F., et al., Ex vivo evaluation of antifibrotic compounds in skin scarring: EGCG and silencing of PAI-1 independently inhibit growth and induce keloid shrinkage. Laboratory investigation, 2013. 93(8): p. 946-960.
    80. Zheng, Y., et al., FePt nanoparticles as a potential X-ray activated chemotherapy agent for HeLa cells. International journal of nanomedicine, 2015. 10: p. 6435.
    81. Mazumder, V., et al., Core/shell Pd/FePt nanoparticles as an active and durable catalyst for the oxygen reduction reaction. Journal of the American Chemical Society, 2010. 132(23): p. 7848-7849.
    82. Donahue, N.D., H. Acar, and S. Wilhelm, Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Advanced drug delivery reviews, 2019. 143: p. 68-96.
    83. Ogiso, T., et al., Effect of positively and negatively charged liposomes on skin permeation of drugs. Journal of drug targeting, 2001. 9(1): p. 49-59.
    84. Yue, Z.-G., et al., Surface charge affects cellular uptake and intracellular trafficking of chitosan-based nanoparticles. Biomacromolecules, 2011. 12(7): p. 2440-2446.
    85. Hirota, K. and H. Terada, Endocytosis of particle formulations by macrophages and its application to clinical treatment. Molecular regulation of endocytosis, 2012. 1.
    86. Sabzevari, A., et al., Polymeric triamcinolone acetonide nanoparticles as a new alternative in the treatment of uveitis: in vitro and in vivo studies. European journal of pharmaceutics and biopharmaceutics, 2013. 84(1): p. 63-71.
    87. Tabatabaei Mirakabad, F.S., et al., PLGA-based nanoparticles as cancer drug delivery systems. Asian Pacific Journal of Cancer Prevention, 2014. 15(2): p. 517-535.
    88. Tan, S., N. Khumalo, and A. Bayat, Understanding keloid pathobiology from a quasi-neoplastic perspective: less of a scar and more of a chronic inflammatory disease with cancer-like tendencies. Frontiers in immunology, 2019. 10: p. 1810.
    89. Vander Heiden, M.G., L.C. Cantley, and C.B. Thompson, Understanding the Warburg effect: the metabolic requirements of cell proliferation. science, 2009. 324(5930): p. 1029-1033.
    90. Xu, J., et al., The radiation therapy in keloids treatment: a comprehensive review of pathomechanism, damage mechanisms and cellular response. 2017.
    91. Moding, E.J., M.B. Kastan, and D.G. Kirsch, Strategies for optimizing the response of cancer and normal tissues to radiation. Nature reviews Drug discovery, 2013. 12(7): p. 526-542.
    92. Schäcke, H., W.-D. Döcke, and K. Asadullah, Mechanisms involved in the side effects of glucocorticoids. Pharmacology & therapeutics, 2002. 96(1): p. 23-43.
    93. De Jong, W.H. and P.J. Borm, Drug delivery and nanoparticles: applications and hazards. International journal of nanomedicine, 2008. 3(2): p. 133.
    94. Shi, Y., et al., Recent advances in FePt nanoparticles for biomedicine. Journal of Nanomaterials, 2015.
    95. Lee, Y.J., et al., Oxidative damage and nuclear factor erythroid 2-related factor 2 protein expression in normal skin and keloid tissue. Annals of dermatology, 2015. 27(5): p. 507-516.
    96. Lingappan, K., NF-κB in oxidative stress. Current opinion in toxicology, 2018. 7: p. 81-86.
    97. Lee, W.J., et al., Decorin‐expressing adenovirus decreases collagen synthesis and upregulates MMP expression in keloid fibroblasts and keloid spheroids. Experimental dermatology, 2015. 24(8): p. 591-597.
    98. Silva, I.R.d., et al., In situ cytokine expression and morphometric evaluation of total collagen and collagens type I and type III in keloid scars. Mediators of inflammation, 2017.
    99. Ji, J., et al., Ionizing irradiation inhibits keloid fibroblast cell proliferation and induces premature cellular senescence. The Journal of dermatology, 2015. 42(1): p. 56-63.
    100. Colell, A., D. Green, and J. Ricci, Novel roles for GAPDH in cell death and carcinogenesis. Cell Death & Differentiation, 2009. 16(12): p. 1573-1581.
    101. Varani, J., Human skin organ culture for assessment of chemically induced skin damage. Expert review of dermatology, 2012. 7(3): p. 295-303.

    無法下載圖示 校內:2025-07-30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