Drug delivery systems for antiangiogenesis and anticancer activities

144 541 0
Drug delivery systems for antiangiogenesis and anticancer activities

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

DRUG DELIVERY SYSTEMS FOR ANTIANGIOGENESIS AND ANTICANCER ACTIVITIES WANG ZHE A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF PHARMACY NATIONAL UNIVERSITY OF SINGAPORE 2010 ACKNOWLEDGEMENTS I am deeply indebted to my main supervisor Associate Professor Ho Chi Lui, Paul and cosupervisor Associate Professor Chui Wai Keung for their continuous encouragement, kind support, careful nurturing and wise advice throughout the period of my PhD study. Sincere appreciation is also extended to Ms. Ng Swee Eng, Ms. Ng Sek Eng, Ms. Quek Mui Hong and Madam Loy Gek Luan for their technical help and support. I would like to thank the Department of Pharmacy, National University of Singapore, for granting me the graduate scholarship to support my study, and for providing me the premises and facilities to carry out the experiments. Additionally, I would like to express my heartfelt gratitude to my dear friends, who never failed to give me great suggestions in both study and life. They are Dr. Lin Haishu, Dr. Huang Meng, Dr. Wu Jinzhu, Dr. Ong Peishi, Dr. Yang Hong, Dr. Anahita Fathi-Azarbayjani, Dr. Wang Chunxia, Dr. Ling Hui, Mr. Li Fang, Mr. Zhang Yaochun, Ms. Cheong Hanhui, Mr. Tarang Nema, Ms. Choo Qiuyi, Ms. Yang Shili, Mr. Sun Feng, Mr. Sun Lingyi. Last but not least, I would like to extend my heartfelt gratitude to my family for their unfailing love and support. I Table of Contents Acknowledgements……………………………………………………………………………… I Table of Contents………………………………………………………………………………….II Summary……………………………………………………………………………………… VIII List of Publications…………………………………………………………………………….…XI List of Tables…………………………………………………………………………………….XII List of Figures………………………………………………………………………………… XIII List of Abbreviations………………………………………………………………………… .XVI Chapter Introduction………………………………………………………………………… .1 1.1 Cancer 1.1.1 Introduction 1.1.2 Metastasis of cancer . 1.1.3 Angiogenesis of cancer . 1.1.4 Relationship of angiogenesis with tumor growth and metastasis . 1.2 Antiangiogenesis therapy 10 1.2.1 Antiangiogenesis inhibitors 10 1.3 Drug delivery system for vascular targeted antiangiogenesis or antitumor therapy 15 1.3.1 Conjugation of drug with ligand for vascular targeting 15 1.3.2 Vascular targeted nanoparticle drug delivery . 19 1.4 Conclusion . 22 Chapter Targeted nanoparticulate drug delivery system to malignant cancer cells 24 2.1 Introduction . 25 2.2 Materials and methods . 26 II 2.2.1 Materials . 26 2.2.2 Conjugation of doxorubicin to PLGA . 27 2.2.3 Preparation of PLGA-doxorubicin (PLGA-Doxo) nanoparticles . 28 2.2.4 Synthesis of Doxo-PLGA-PEG-cRGD nanoparticles 28 2.2.5 Characterization of nanoparticles . 29 2.2.6 Cytotoxicity assay 29 2.2.7 Cell uptake and binding affinity assays 30 2.2.8 DNA fragmentation assay 30 2.2.9 Western blot analysis 31 2.2.10 In vitro drug release 32 2.3 Results . 32 2.3.1 Conjugation of doxorubicin to PLGA 32 2.3.2 Synthesis and characterization of Doxo-PLGA-PEG-cRGD nanoparticle . 33 2.3.3 Cytotoxicity of NPs to various malignant cancer cells . 35 2.3.4 Cell affinity assay . 36 2.3.5 In vitro drug release 37 2.3.6 Cell apoptosis induced by Doxo-PLGA-PEG-cRGD NPs . 37 2.4 Discussion . 39 2.5 Conclusion . 43 Chapter In vitro evaluation of vascular targeted nanocapsule with sequential drug delivery for temporal antiendothelial and anticancer activities 44 3.1 Introduction . 45 III 3.2 Materials and methods . 46 3.2.1. Preparation of Paclitaxel loaded PLGA core . 46 3.2.2. Preparation of core-shell nanocapsule . 47 3.2.3. Conjugation of RGDfK peptide to the nanocapsule 48 3.2.4. In vitro drug release from the core-shell nanocapsule . 48 3.2.5. Cell apoptosis assay . 48 3.2.6. Immunofluorescence staining of tubulin disruption 49 3.2.7. Cellular uptake of the nanocapsule 49 3.2.8. Confocal laser scanning microscope (CLSM) observation of the fluorophores labeled nanocapsule . 50 3.2.9 Co-culture assay . 50 3.3 Results and discussion . 51 3.4 Conclusion . 61 Chapter Targeted nanoparticle drug delivery for combined antiangiogenesis and antitumor activities…………………………………………………………………………… .62 4.1 Introduction . 63 4.2 Materials and methods . 64 4.2.1 Material 64 4.2.1 Preparation of drug loaded nanoparticles . 65 4.2.2 Characterization of nanoparticles . 66 4.2.3 Cellular uptake of NPs and their distribution in cells . 67 4.2.4 Western blot to detect cellular apoptosis induced by PTX NPs . 68 IV 4.2.5 Immnunofluorescence staining of tubulin 68 4.2.6 In vivo antitumor studies 69 4.2.7 Immunohistochemical staining . 70 4.3 Results . 70 4.3.1 Preparation and characterization of targeted nanoparticles 70 4.3.2 Cellular uptake of nanoparticles . 72 4.3.3 In vitro evaluation of efficacy of nanoparticles 75 4.3.4 Antitumor evaluation of different formulations . 77 4.3.5 Histological evaluation of different formulation treatment 78 4.4 Discussion . 80 4.5 Conclusion . 84 Chapter Sequential drug delivery by nanocapsule for targeted disruption of tumor vasculature to enhance anticancer and antimetastatic activities…………………………… 85 5.1 Introduction . 86 5.2 Materials and methods . 87 5.2.1Material . 87 5.2.2 Cell lines and animals. 88 5.3.3 Drug conjugation and nanocapsule preparation method 88 5.2.4 Characterization of nanocapsule . 89 5.2.5 Uptake of nanocapsule by HUVECs and intracellular distribution in cells . 89 5.2.6 Annexin V/propidium iodide apoptosis staining assay 90 5.2.7 Scratch assay 90 V 5.2.8 Tube formation assay . 91 5.2.9 Body distribution and toxicity studies of nanocapsule . 91 5.2.10 In vivo Matrigel plug assay . 92 5.2.11 In vivo primary tumor experiment 92 5.2.12 Liver metastasis prevention experiment . 93 5.2.13 Immunohistochemistry evaluation of angiogenesis, proliferation and apoptosis in tumor . 93 5.3 Results . 93 5.3.1 Physicochemical characterization of nanocapsule 93 5.3.2 Nanocapsule uptake by endothelial cells through endocytosis . 95 5.3.3 Sustained release of conjugated paclitaxel from nanocapsule induced apoptosis in cancer cells 95 5.3.4 The potency of CA4 loaded nanocapsule in inhibiting HUVEC proliferation and initiating vascular disruption effect in vitro 98 5.3.5 Biodistribution, tumor accumulation and tissue toxicity studies of nanocapsule . 99 5.3.6 Antiangiogenesis effect of nanocapsule in the Matrigel® plug model. . 101 5.3.7 Antivasculature and primary tumor growth inhibition effects of nanocapsule 101 5.3. Liver metastasis prevention of nanocapsule. . 107 5.4 Discussion . 108 5.5 Conclusion . 111 Chapter Conclusion and Future Direction…………………………………………………112 6.1 Conclusion . 113 VI 6.2 Future Direction 116 BIBLIOGRAPY……………………………………………………………………… .…… .117 VII Summary Angiogenesis is one of the vital events for organ development and differentiation during embryogenesis as well as wound healing and reproductive functions in adults. Angiogenesis also contributes significantly to tumor growth and metastasis. Hence, search for effective antiangiogenesis agents and therapy is an emerging field in the past decades. From the existing experience, the mono-antiangiogenesis therapy, however, is hampered by the fact that cancer cells could evade the single antiangiogenesis treatment designated to control tumor endothelial cell growth and tumor cell survival. Therefore, combinatorial therapy in which antiangiogenetic agent and chemotherapeutic drug administrated in a scheduled manner could result in a much more favorable therapeutic effect. In this thesis, we designed and fabricated several multifunctional and/or combinatorial nanoparticulated delivery systems aiming to disrupt tumor endothelial cells and induce apoptosis in tumor cells simultaneously for antiangiogenesis and anticancer therapy. To realize the specific tumor site targeting ability, we used a RGD (Arg-Gly-Asp) sequence containing peptide as the targeting ligand, which could target not only to the universally available endothelial cells in tumor microenvironment, but also to some specific types of malignant cancer cells via the overexpressed integrin αvβ3 and/or αvβ5 on cell membrane. In our first study, we investigated the potency of our designed doxorubicin conjugated nanoparticle decorated with PEG (poly(ethylene glycol)) and RGD peptide on surface for drug delivery and apoptosis induction in three malignant cancer cells, DU145, MDA-MB-231, and B16F10. All the three cancer cells were reported to express integrin αvβ3 and/or αvβ5 on cell membrane in different extents. Results showed that this drug conjugated targeted delivery system could be taken up by malignant cells efficiently; and its apoptosis induction ability on particular cancer cells was also effective, even though the overall VIII therapeutic efficacy of the conjugated anticancer drug was, to some extent, compromised after conjugation. In addition, the drug release profile of doxorubin from the matrix was in a sustained release manner (zero-order, r2 > 0.97). In our second study, we prepared a RGD modified core-shell nanocapsule delivery system to investigate the capacity of temporal release of antiangiogenetic drug and anticancer drug in vitro. We found that the drug release profiles of the encapsulated drugs, paclitaxel (PTX) and combretastatin (CA4) could be well-controlled by changing some preparation parameters, such as the density of phospholipid or matrix materials fed. We also found that the uptake of this nanocapsule by the endothelial cells was efficient, and the endothelial cellular cytoskeleton could be effectively disrupted by the antiangiogenetic agent, CA4. The PTX loaded in the nanoparticle core still demonstrated effective therapeutic effect to cancer cells. What is more important, our results indicated that the sequential release of CA4 and PTX from the nanocapsule could temporally diminish endothelial cells before eliminating the cancer cells with the potential to shut down the tumor microvasculature and trap the anticancer drug inside the tumor. Thus, it was evidenced that antiangiogenesis and anticancer activities could be successfully realized with appropriate formulation design and material choice. In our third study, we employed a xenograft tumor bearing mouse model to evaluate our nanoparticulate delivery system for combinatorial therapy. PTX and CA4 were co-formulated into the RGD decorated nanoparticle in a robust nanoprecipitation method. Results from the in vivo model showed that malignant tumor growth was significantly suppressed with this combinatorial method, and histological examination revealed that the mechanism underlining this encouraging therapeutic efficacy was the disruption of tumor vasculature and tumor apoptosis induction by the combined drugs. To establish the hypothesis on the advantage of having temporal delivery of antiangiogenesis and anticancer drugs with our nanoparticulate delivery system, we further designed a micelle delivery system without RGD peptide decorated on the IX polymer is a critical factor in determining the release and efficacy of polymer conjugated drugs (83, 194). The large molecular weight of PLGA (61kDa) used by Sengupta et al. may delay the hydrolysis of the polymer-drug conjugate, thus leading to compromised anticancer drug efficacy. The molecular weight of PLA used for our nanoparticle was optimized at KDa. It could lead to efficient hydrolysis of the conjugate in the cytoplasm. In addition, the conjugation of PLGA to the 3’-NH2 group of doxorubicin via amide bond in their study did not release doxorubicin in the original drug form but as the Doxo-3’-lactamide prodrug (195), leading to compromised therapeutic effect when used alone. In contrast, in our nanocapsule, the 2’-OH of paclitaxel was conjugated to the carboxyl end of PLA via ester bond that could be easily cleaved by hydrolysis upon cellular uptake (177). By choosing a suitable polymer and optimizing the molecular weight of the build-up polyester and the synthetic route for the polymer-drug conjugation, effective drug loading and sequential release of drugs were achieved with our novel nanocapsule. 5.5 Conclusion We have fabricated a novel combinatorial drug delivery nanocapsule with sequential delivery feature for effective antivasculature and anticancer activities in vitro and in vivo. The findings in this study could encourage further investigation of other combinatorial treatments administered in the same or similar delivery systems for effective malignant cancer therapy. In addition, this novel nanocapsule delivery system could be employed as a template for other applications, such as by adding iron oxide nanoparticle in the core of the system for the simultaneous non-invasive in vivo imaging. 111 Chapter Conclusion and Future Direction 112 6.1 Conclusion In this thesis, we investigated combinatorial drug delivery by nanoparticulated particle for simultaneous antiangiogenesis and anticancer therapy. With the rapid advancing of nanotechnology, nanoparticulated drug delivery systems play important roles in biomedical applications, including cancer therapy. Nanoparticles loaded with therapeutic agents could extend the circulation time of drug in human body, accumulate anticancer drug at specific tumor sites, and alleviate the potential side-effects of drugs. Therefore, many investigations have focused on the novel drug delivery systems from synthesizing basic materials components to delivery system fabrication methods. Nevertheless, few efforts have been paid on the therapeutic effects of drug delivery systems on the malignant metastatic cancers. In the metastatic progress of tumor cells, angiogenesis in tumor microenvironment is a key player for tumor malignance and widespreading in the entire body. Thus, the antiangiogenesis therapy was popularly explored, ever since this concept was proposed by Folkman in 1971 (40). Along with the better understanding of biological process of angiogenesis and corresponding discovery and development of antiangiogenic agents, numerous lines of evidences have proved that antiangiogenic agents as monotherapeutics have limited, if any, discernable efficacy, especially in the treatment of advanced malignant cancers. On the other hand, the conventional chemotherapy can result in the well-known drug resistance effect after long-term administration. This drug resistance sets a barrier for better treatment of cancer. In addition, due to the plasticity, tumors are likely to evade single targeted therapeutic method designated to control the proliferation and survival of tumor cells. Although the developed drug delivery systems could, to some extent, alleviated the drug resistance effects of many anticancer drugs and deliver drug in high pay-load to tumor sites, as well as diminishing the side-effects, more effective combination therapy drug delivery systems with simultaneous antiangiogenesis and anticancer activities are urgently desired. 113 Herein, we hypothesize that drug delivery system under controllable drug release features with combined antiangiogenesis and anticancer agents could more efficiently and effectively suppress the growth of malignant tumor masses, decrease the angiogenesis progress, and prevent the metastasis of such tumors in cancer therapy. In chapter of this thesis, a small peptides containing Arg-Gly-Asp (RGD) sequence was selected as the targeting moiety. This RGD sequence was reported as a highly affinitive ligand to αvβ3 and αvβ5 receptors overexpressed on many cancerous cells (83) and highly proliferating tumor endothelial cells (151). In this chapter, we first investigated the potency of RGD surface modified nanoparticle with tethering PEG for delivery of conjugated doxorubicin to different types of malignant metastatic cells. Experiments had demonstrated that the synthesized NPs in this study were able to effectively target conjugated doxorubicin to malignant integrin expression cancer cells, and showed profound apoptotic induction effect to those cells. Although the drug efficacy to cancer cells was compromised after conjugation with PLGA, the resultant sustained release behavior and the property of selective uptake of the NPs by cancer cells could still make Doxo-PLGA-PEG-cRGD NPs a good candidate for anticancer therapy. In chapter 3, we explored the endothelial cell targeting ability of RGD surface tagged nanocapsule with temporal antiangiogenesis and anticancer drug release feature by using a robust self-assembly nanoprecipitation method. The combinatorial nanocapsule preserved the high cytotoxic effect of the anticancer drug encapsulated in the core, and could deliver the drugs in a controlled sustained manner to disrupt tumor vasculature and ablate tumor cells sequentially with the assistance of RGD targeting ligand. Results from chapter and chapter evidenced that these targeted nanoparticulated delivery systems hold the ability for effective anticancer or simultaneous antiangiogenesis and anticancer therapy. 114 In chapter 4, we encapsulated two drugs, PTX and CA4 for the respective anticancer and antiangiogenesis effects, into a RGD targeted nanoparticle, the matrix of which was composed of biodegradable PLGA polyester. The cellular uptake of the prepared nanoparticle was efficient. The nanoparticle loaded with PTX could efficiently induce apoptosis in cancer cells, and disrupt the cytoskeleton when loaded with CA4, respectively. Xenograft tumor model confirmed the effective therapeutic effects of targeted dual-drug loaded nanoparticle, and histological tests also established the mechanism of efficacy by inducing cancer cell apoptosis and tumor endothelial cell death. Therefore, we envision that this robust targeted nanoparticle with combined therapy for antiangiogenesis and anticancer activities may show a formidable candidate for cancer therapy. In chapter 5, we further explored our concept and applied with a more advanced and a welldefined micellar delivery system, in which the polyester was conjugated to PTX to form a core for the controlled and sustained anticancer drug delivery, The polyester-PTX conjugate was further used to physically encapsulate CA4 for the antiangiogenesis function. Interestingly, we found that without RGD targeting function, micelles with appropriate particle size and PEG surface tethering could be uptaken by endothelial cells efficiently, and induce cytoskeleton disruption effectively upon fast CA4 releasing. The drug release profile could also be adjusted accordingly by conjugating the drug to the polyester with different molecular weights, and by altering the encapsulation parameters. The biodistribution experiments demonstrated the long circulation of nanocapsule in body fluid and the preferential accumulation of nanocapsule in tumor. Both in vivo artificial pro-angiogenesis and tumor xenograft assays demonstrated the promising therapeutic effect of the nanocapsule on tumor vasculature disruption, tumor cell proliferation inhibition and tumor cell apoptosis induction. The intrasplenic liver metastasis experiment also confirmed the liver metastatic prevention capacity of this novel nanocapsule. In summary, the findings indicated that this novel dual drug loaded micelle with sequential drug 115 delivery capacity is a promising candidate in combinatorial therapy in fighting against cancer, and may open an avenue for cancer therapy and diagnosis. In summary, this thesis demonstrated the in vitro and/or in vivo efficacy of several prototypes of nanoparticulated delivery systems for combinatorial temporal drug delivery for antiangiogenesis and anticancer therapy. The fabrication of these systems is robust and simple and can be easily modified for the other combinatorial temporal drug delivery. Last but not least, we hope that our study could, to some degree, contribute to the advance of cancer research for better healthcare. 6.2 Future Direction For the future study of this project, there are still some important issues to be addressed: firstly, the different pharmaceutical formulations developed in this study need to be further optimized to enhance the drug loading efficiency and encapsulation efficiency and provide high nanoparticle stability; secondly, other promising tumor vasculature and/or cancerous cell surface receptor targeting ligands such as antibodies or small molecules other than the RGD sequence containing peptide widely used in this study could be utilized for the active targeting purpose; thirdly, some imaging contrast agents could be co-encapsulated in the appropriate nanoparticle formulation for simultaneous bioimaging to monitor the therapeutic function arising from the drugs; fourthly, other tumor animal models could be used to further evaluate the therapeutic efficacy of these delivery systems in cancer treatment. In addition, a broader range of tumor types, such as prostate cancer, breast cancer, brain cancer, may be adopted to establish the versatile therapeutic effects of the developed nanoparticles. The application of these systems for gene therapy, such as siRNA for specific mutant gene silencing, coupled with antiangiogenesis agents could also be explored. 116 BIBLIOGRAPY 1. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006. Ca-a Cancer Journal for Clinicians 2006;56:106-30. 2. Renan MJ. How many mutations are required for tumorigenesis - Implications from human cancer data. Molecular Carcinogenesis 1993;7:139-46. 3. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70. 4. Page DL, Kidd TE, Dupont WD, Simpson JF, Rogers LW. Lobular neoplasia of the breast - higher risk for subsequent invasive cancer predicted by more extensive disease. Human Pathology 1991;22:1232-9. 5. Weber WP, Zanetti R, Langer I, et al. Mammotome: Less invasive than ABBI with similar accuracy for early breast cancer detection. World Journal of Surgery 2005;29:495-9. 6. Jemal A, Siegel R, Ward E, Murray T, Xu JQ, Thun MJ. Cancer statistics, 2007. Ca-a Cancer Journal for Clinicians 2007;57:43-66. 7. Merlo LMF, Pepper JW, Reid BJ, Maley CC. Cancer as an evolutionary and ecological process. Nature Reviews Cancer 2006;6:924-35. 8. Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Research 1975;35:512-6. 9. Fidler IJ. Metastasis - Quantitative analysis of distribution and fate of tumor emboli labled with I-125-5-iodo-2'-deoxyruidine. Journal of the National Cancer Institute 1970;45:773-&. 10. Wyckoff JB, Jones JG, Condeelis JS, Segall JE. A critical step in metastasis: In vivo analysis of intravasation at the primary tumor. Cancer Research 2000;60:2504-11. 11. Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005;438:820-7. 12. Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer metastasis reviews 1989;8:98-101. 13. James. E, editor. Neoplastic Diseases. A Treatise on Tumors: Philadelphia London, W. B. Saunders company; 1928. 14. Hart IR, Fidler IJ. Role of organ selectivity in the determination of metastatic patterns of B-16 melanoma. Cancer Research 1980;40:2281-7. 15. Weiss L, Harlos JP. The validity of negative necropsy reports for metastases in solid organs. Journal of Pathology 1986;148:203-6. 16. Weiss L, Grundmann E, Torhorst J, et al. Haematogenous metastatic patterns in colonic carcinoma: an analysis of 1541 necropsies. The Journal of pathology 1986;150:195-203. 17. Weiss L. Comments on hematogenous metastatic patterns in humans as revealed by autopsy. Clinical & Experimental Metastasis 1992;10:191-9. 18. Welch DR. Technical considerations for studying cancer metastasis in vivo. Clinical & Experimental Metastasis 1997;15:272-306. 19. Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nature Medicine 2006;12:895-904. 20. Folkman J. Aangiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine 1995;1:27-31. 21. Salven P, Mustjoki S, Alitalo R, Alitalo K, Rafii S. VEGFR-3 and CD133 identify a population of CD34(+) lymphatic/vascular endothelial precursor cells. Blood 2003;101:168-72. 22. Battegay EJ. Angiogenesis - mechanistic insights, neovascular diseases, and therapeutic prospects. Journal of Molecular Medicine-Jmm 1995;73:333-46. 23. Nyberg P, Xie L, Kalluri R. Endogenous inhibitors of angiogenesis. Cancer Research 2005;65:3967-79. 117 24. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353-64. 25. Bouck N, Stellmach V, Hsu SC. How tumors become angiogenic. Advances in Cancer Research, Vol 69; 1996. p. 135-74. 26. Sunderkotter C, Goebeler M, Schulzeosthoff K, Bhardwaj R, Sorg C. Macrophagederived angiogenesis factors. Pharmacology & Therapeutics 1991;51:195-216. 27. Hanahan D. Signaling vascular morphogenesis and maintenance. Science 1997;277:48-50. 28. Conway EM, Collen D, Carmeliet P. Molecular mechanisms of blood vessel growth. Cardiovascular Research 2001;49:507-21. 29. Auerbach R, Kubai L, Sidky Y. Angiogenesis induction by tumors, embryonic tissues, and lymphocytes. Cancer Research 1976;36:3435-40. 30. Hori K, Suzuki M, Tanda S, Saito S. In vivo analysis of tumor vascularization in the rat. Jpn J Cancer Res 1990;81:279-88. 31. Dvorak HF, Nagy JA, Dvorak AM. Structure of solid tumors and theri vasculature implications for therapy with monoclone-antibodies. Cancer Cells-a Monthly Review 1991;3:7785. 32. Campbell SC. Advances in angiogenesis research: Relevance to urological oncology. Journal of Urology 1997;158:1663-74. 33. Folkman J. The role of angiogenesis in tumor growth. Seminars in cancer biology 1992;3:65-71. 34. Gimbrone MA, Cotran RS, Folkman J, Leapman SB. Tumor dormancy in vivo by prevention of neovascularization. Journal of Experimental Medicine 1972;136:261-&. 35. Kim KJ, Li B, Winer J, et al. Inhibition of vascular endothelial growth factor induced angiogenesis suppresses tumor growth in vivo. Nature 1993;362:841-4. 36. Hori A, Sasada R, Matsutani E, et al. Suppsression of solid tumor growth by immunoneutralizing monoclonal antibody against human basic fibroblast growth factor. Cancer Research 1991;51:6180-4. 37. Liotta LA, Kleinerm.J, Saidel GM. Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation. Cancer Research 1974;34:997-1004. 38. Oreilly MS, Holmgren L, Chen C, Folkman J. Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Medicine 1996;2:689-92. 39. Weidner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis correlation in invasive breast carcinoma. New England Journal of Medicine 1991;324:1-8. 40. Folkman J. Tumor angiogenesis: therapeutic implications. The New England journal of medicine 1971;285:1182-6. 41. Ingber D, Fujita T, Kishimoto S, et al. Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumour growth. Nature 1990;348:555-7. 42. Castronovo V, Belotti D. TNP-470 (AGM-1470): mechanisms of action and early clinical development. Eur J Cancer 1996;32A:2520-7. 43. Rastinejad F, Polverini PJ, Bouck NP. Regulation of the activity of a new inhibitor of angiogenesis by a cancer suppressor gene. Cell 1989;56:345-55. 44. Luster AD, Greenberg SM, Leder P. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor and inhibits endothelial cell proliferation. The Journal of experimental medicine 1995;182:219-31. 45. Oreilly MS, Holmgren L, Shing Y, et al. Angiostatin - A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis Lung Carcinoma. Cell 1994;79:315-28. 46. Dhanabal M, Ramchandran R, Waterman MJF, et al. Endostatin induces endothelial cell apoptosis. Journal of Biological Chemistry 1999;274:11721-6. 118 47. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nature Reviews Drug Discovery 2004;3:391-400. 48. Wildiers H, Guetens G, De Boeck G, et al. Effect of antivascular endothelial growth factor treatment on the intratumoral uptake of CPT-II. British Journal of Cancer 2003;88:1979-86. 49. Fong TAT, Shawver LK, Sun L, et al. SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Research 1999;59:99-106. 50. Potapova O, Laird AD, Nannini MA, et al. Contribution of individual targets to the antitumor efficacy of the multitargeted receptor tyrosine kinase inhibitor SU11248. Mol Cancer Ther 2006;5:1280-9. 51. Broutyboye D, Zetter BR. Inhibition of cell motility by interferon. Science 1980;208:5168. 52. Sidky YA, Borden EC. Inhibition of angiogenesis by interferons - effects on tumorinduced and lymphocyte-induced vascular responses. Cancer Research 1987;47:5155-61. 53. Ezekowitz RAB, Mulliken JB, Folkman J. Inteferon alfa-2a therapy for life-threatening hemangiomas of infancy. New England Journal of Medicine 1992;326:1456-63. 54. Hynes RO. Integrins: Bidirectional, allosteric signaling machines. Cell 2002;110:673-87. 55. Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nature Reviews Cancer 2002;2:91-+. 56. Burridge K, ChrzanowskaWodnicka M. Focal adhesions, contractility, and signaling. Annual Review of Cell and Developmental Biology 1996;12:463-518. 57. Wang Z, Chui WK, Ho PC. Integrin targeted drug and gene delivery. Expert Opinion on Drug Delivery;7:159-71. 58. Cheresh DA. Human-Endothelial Cells Synthesize and Express an Arg-Gly-Asp-Directed Adhesion Receptor Involved in Attachment to Fibrinogen and Vonwillebrand-Factor. Proceedings of the National Academy of Sciences of the United States of America 1987;84:64715. 59. Chen QM, Manning CD, Millar H, et al. CNTO 95, a fully human anti alpha v integrin antibody, inhibits cell signaling, migration, invasion, and spontaneous metastasis of human breast cancer cells. Clinical & Experimental Metastasis 2008;25:139-48. 60. Dechantsreiter MA, Planker E, Matha B, et al. N-methylated cyclic RGD peptides as highly active and selective alpha(v)beta(3) integrin antagonists. Journal of Medicinal Chemistry 1999;42:3033-40. 61. Khalili P, Arakelian A, Chen GP, et al. A non-RGD-based integrin binding peptide (ATN-161) blocks breast cancer growth and metastasis in vivo. Mol Cancer Ther 2006;5:2271-80. 62. Thibault G, Tardif P, Lapalme G. Comparative specificity of platelet alpha(IIb)beta(3) integrin antagonists. Journal of Pharmacology and Experimental Therapeutics 2001;296:690-6. 63. Griffioen AW, Molema G. Angiogenesis: Potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation. Pharmacological Reviews 2000;52:237-68. 64. Rasmussen HS, McCann PP. Matrix metalloproteinase inhibition as a novel anticancer strategy: A review with special focus on batimastat and marimastat. Pharmacology & Therapeutics 1997;75:69-75. 65. Brown PD, Giavazzi R. Matrix metalloproteinase inhibition: A review of anti-tumour activity. Annals of Oncology 1995;6:967-74. 66. Brooks PC, Silletti S, von Schalscha TL, Friedlander M, Cheresh DA. Disruption of angiogenesis by PEX, a noncatalytic metalloproteinase fragment with integrin binding activity. Cell 1998;92:391-400. 67. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nature Medicine 2001;7:987-9. 119 68. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 1998;279:377-80. 69. Kim JW, Lee HS. Tumor targeting by doxorubicin-RGD-4C peptide conjugate in an orthotopic mouse hepatoma model. International Journal of Molecular Medicine 2004;14:529-35. 70. Burkhart DJ, Kalet BT, Coleman MP, Post GC, Koch TH. Doxorubicin-formaldehyde conjugates targeting alpha(v)beta(3) integrin. Mol Cancer Ther 2004;3:1593-604. 71. Chen XY, Plasencia C, Hou YP, Neamati N. Synthesis and biological evaluation of dimeric RGD peptide-paclitaxel conjugate as a model for integrin-targeted drug delivery. Journal of Medicinal Chemistry 2005;48:1098-106. 72. Cao QZ, Li ZB, Chen K, et al. Evaluation of biodistribution and anti-tumor effect of a dimeric RGD peptide-paclitaxel conjugate in mice with breast cancer. European Journal of Nuclear Medicine and Molecular Imaging 2008;35:1489-98. 73. Dunehoo AL, Anderson M, Majumdar S, Kobayashi N, Berkland C, Siahaan TJ. Cell adhesion molecules for targeted drug delivery. Journal of Pharmaceutical Sciences 2006;95:185672. 74. Ellerby HM, Arap W, Ellerby LM, et al. Anti-cancer activity of targeted pro-apoptotic peptides. Nature Medicine 1999;5:1032-8. 75. Chen YX, Xu XM, Hong SG, et al. RGD-tachyplesin inhibits tumor growth. Cancer Research 2001;61:2434-8. 76. Yokoyama Y, Ramakrishnan S. Addition of integrin binding sequence to a mutant human endostatin improves inhibition of tumor growth. International Journal of Cancer 2004;111:839-48. 77. Curnis F, Gasparri A, Sacchi A, Longhi R, Corti A. Coupling tumor necrosis factor-alpha with alpha(v) integrin ligands improves its antineoplastic activity. Cancer Research 2004;64:56571. 78. Sacchi A, Gasparri A, Curnis F, Bellone M, Corti A. Crucial role for interferon gamma in the synergism between tumor vasculature-targeted tumor necrosis factor alpha (NGR-TNF) and doxorubicin. Cancer Research 2004;64:7150-5. 79. Xiao B, Li W, Yang J, Guo G, Mao XH, Zou QM. RGD-IL-24, A Novel Tumor-Targeted Fusion Cytokine: Expression, Purification and Functional Evaluation. Molecular Biotechnology 2009;41:138-44. 80. Bieker R, Kessler T, Schwoppe C, et al. Infarction of tumor vessels by NGR-peptidedirected targeting of tissue factor: experimental results and first-in-man experience. Blood 2009;113:5019-27. 81. Dickerson EB, Akhtar N, Steinberg H, et al. Enhancement of the antiangiogenic activity of interleukin-12 by peptide targeted delivery of the cytokine to alpha(v)beta(3) integrin. Mol Cancer Res 2004;2:663-73. 82. Matsumura Y, Maeda H. A New Concept for Macromolecular Therapeutics in CancerChemotherapy - Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Research 1986;46:6387-92. 83. Wang Z, Chui WK, Ho PC. Design of a Multifunctional PLGA Nanoparticulate Drug Delivery System: Evaluation of its Physicochemical Properties and Anticancer Activity to Malignant Cancer Cells. Pharmaceutical Research 2009;26:1162-71. 84. Murphy EA, Majeti BK, Barnes LA, et al. Nanoparticle-mediated drug delivery to tumor vasculature suppresses metastasis. Proceedings of the National Academy of Sciences of the United States of America 2008;105:9343-8. 85. Xiong XB, Huang Y, Lu WL, et al. Enhanced intracellular delivery and improved antitumor efficacy of doxorubicin by sterically stabilized liposomes modified with a synthetic RGD mimetic. Journal of Controlled Release 2005;107:262-75. 86. Xiong XB, Huang Y, Lu WL, et al. Intracellular delivery of doxorubicin with RGDmodified sterically stabilized liposomes for an improved antitumor efficacy: In vitro and in vivo. Journal of Pharmaceutical Sciences 2005;94:1782-93. 120 87. Dubey PK, Mishra V, Jain S, Mahor S, Vyas SP. Liposomes modified with cyclic RGD peptide for tumor targeting. Journal of Drug Targeting 2004;12:257-64. 88. Holig P, Bach M, Volkel T, et al. Novel RGD lipopeptides for the targeting of liposomes to integrin-expressing endothelial and melanoma cells. Protein Engineering Design & Selection 2004;17:433-41. 89. Zhao H, Wang JC, Sun QS, Luo CL, Zhang Q. RGD-based strategies for improving antitumor activity of paclitaxel-loaded liposomes in nude mice xenografted with human ovarian cancer. Journal of Drug Targeting 2009;17:10-8. 90. Pattillo CB, Sari-Sarraf F, Nallamothu R, Moore BM, Wood GC, Kiani MF. Targeting of the antivascular drug combretastatin to irradiated tumors results in tumor growth delay. Pharmaceutical Research 2005;22:1117-20. 91. Sen Gupta A, Huang G, Lestini BJ, Sagnella S, Kottke-Marchant K, Marchant RE. RGDmodified liposomes targeted to activated platelets as a potential vascular drug delivery system. Thrombosis and Haemostasis 2005;93:106-14. 92. Lestini BJ, Sagnella SM, Xu Z, et al. Surface modification of liposomes for selective cell targeting in cardiovascular drug delivery. Journal of Controlled Release 2002;78:235-47. 93. Nasongkla N, Shuai X, Ai H, et al. cRGD-functionalized polymer micelles for targeted doxorubicin delivery. Angewandte Chemie-International Edition 2004;43:6323-7. 94. Nasongkla N, Bey E, Ren JM, et al. Multifunctional polymeric micelles as cancertargeted, MRI-ultrasensitive drug delivery systems. Nano Letters 2006;6:2427-30. 95. Deng C, Chen XS, Sun J, Lu TC, Wang WS, Jing XB. RGD peptide grafted biodegradable amphiphilic triblock copolymer poly(glutamic acid)-b-poly(L-lactide)-bpoly(glutamic acid): Synthesis and self-assembly. Journal of Polymer Science Part a-Polymer Chemistry 2007;45:3218-30. 96. Shen SI, Kotamraj PR, Bhattacharya S, Li XL, Jasti BR. Synthesis and characterization of RGD-fatty acid amphiphilic micelles as targeted delivery carriers for anticancer agents. Journal of Drug Targeting 2007;15:51-8. 97. Kim JH, Kim YS, Park K, et al. Self-assembled glycol chitosan nanoparticles for the sustained and prolonged delivery of antiangiogenic small peptide drugs in cancer therapy. Biomaterials 2008;29:1920-30. 98. Swenson S, Costa F, Minea R, et al. Intravenous liposomal delivery of the snake venom disintegrin contortrostatin limits breast cancer progression. Mol Cancer Ther 2004;3:499-511. 99. Schiffelers RM, Koning GA, ten Hagen TLM, et al. Anti-tumor efficacy of tumor vasculature-targeted liposomal doxorubicin. Journal of Controlled Release 2003;91:115-22. 100. Schiffelers RM, Ansari A, Xu J, et al. Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle. Nucleic Acids Research 2004;32. 101. Temming K, Schiffelers RM, Molema G, Kok RJ. RGD-based strategies for selective delivery of therapeutics and imaging agents to the tumour vasculature. Drug Resist Update 2005;8:381-402. 102. Langer R. Perspectives: Drug delivery - Drugs on target. Science 2001;293:58-9. 103. Davis ME, Chen Z, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nature Reviews Drug Discovery 2008;7:771-82. 104. Lammers T, Hennink WE, Storm G. Tumour-targeted nanomedicines: principles and practice. British Journal of Cancer 2008;99:392-7. 105. Dandamudi S, Campbell RB. The drug loading, cytotoxicty and tumor vascular targeting characteristics of magnetite in magnetic drug targeting. Biomaterials 2007;28:4673-83. 106. Papaetis GS, Roussos C, Syrigos KN. Targeted therapies for non-small cell lung cancer. Current Pharmaceutical Design 2007;13:2810-31. 107. Bae Y, Nishiyama N, Kataoka K. In vivo antitumor activity of the folate-conjugated pHSensitive polymeric micelle selectively releasing adriamycin in the intracellular acidic compartments. Bioconjugate Chemistry 2007;18:1131-9. 121 108. Peer D, Karp JM, Hong S, FaroKhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nature Nanotechnology 2007;2:751-60. 109. Yoo HS, Lee KH, Oh JE, Park TG. In vitro and in vivo anti-tumor activities of nanoparticles based on doxorubicin-PLGA conjugates. J Control Release 2000;68:419-31. 110. Oh JE, Nam YS, Lee KH, Park TG. Conjugation of drug to poly(D,L-lactic-co-glycolic acid) for controlled release from biodegradable microspheres. Journal of Controlled Release 1999;57:269-80. 111. Yoo HS, Oh JE, Lee KH, Park TG. Biodegradable nanoparticles containing doxorubicinPLGA conjugate for sustained release. Pharm Res 1999;16:1114-8. 112. Zhang N, Chittasupho C, Duangrat C, Siahaan TJ, Berkland C. PLGA nanoparticle-peptide conjugate effectively targets intercellular cell-adhesion molecule-1. Bioconjug Chem 2008;19:145-52. 113. Chen F, Liu Y, Lu J, Hwang KJ, Lee VH. A sensitive fluorometric assay for reducing sugars. Life Sci 1992;50:651-9. 114. Herrmann M, Lorenz HM, Voll R, Grunke M, Woith W, Kalden JR. A rapid and simple method for the isolation of apoptotic DNA fragments. Nucleic Acids Res 1994;22:5506-7. 115. Keegan ME, Royce SM, Fahmy T, Saltzman WM. In vitro evaluation of biodegradable microspheres with surface-bound ligands. J Control Release 2006;110:574-80. 116. Vellon L, Menendez JA, Liu H, Lupu R. Up-regulation of alphavbeta3 integrin expression is a novel molecular response to chemotherapy-induced cell damage in a heregulindependent manner. Differentiation 2007;75:819-30. 117. Ramos OH, Kauskot A, Cominetti MR, et al. A novel alpha(v)beta (3)-blocking disintegrin containing the RGD motive, DisBa-01, inhibits bFGF-induced angiogenesis and melanoma metastasis. Clin Exp Metastasis 2008;25:53-64. 118. Line BR, Mitra A, Nan A, Ghandehari H. Targeting tumor angiogenesis: Ccomparison of peptide and polymer-peptide conjugates. Journal of Nuclear Medicine 2005;46:1552-60. 119. Yang RS, Tang CH, Chuang WJ, et al. Inhibition of tumor formation by snake venom disintegrin. Toxicon 2005;45:661-9. 120. Cai W, Shin DW, Chen K, et al. Peptide-labeled near-infrared quantum dots for imaging tumor vasculature in living subjects. Nano Lett 2006;6:669-76. 121. Pattillo CB, Sari-Sarraf F, Nallamothu R, Moore BM, Wood GC, Kiani MF. Targeting of the antivascular drug combretastatin to irradiated tumors results in tumor growth delay. Pharm Res 2005;22:1117-20. 122. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 1998;279:377-80. 123. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 2004;56:185-229. 124. Meyer-Losic F, Quinonero J, Dubois V, et al. Improved therapeutic efficacy of doxorubicin through conjugation with a novel peptide drug delivery technology (Vectocell). J Med Chem 2006;49:6908-16. 125. Rihova B, Kubackova K. Clinical implications of N-(2-hydroxypropyl)methacrylamide copolymers. Current Pharmaceutical Biotechnology 2003;4:311-22. 126. Lammers T, Subr V, Peschke P, et al. Image-guided and passively tumour-targeted polymeric nanomedicines for radiochemotherapy. Br J Cancer 2008;99:900-10. 127. Lammers T, Peschke P, Kuehnlein R, et al. Effect of intratumoral injection on the biodistribution and the therapeutic potential of HPMA copolymer-based drug delivery systems. Neoplasia 2006;8:788-95. 128. Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer 2006;6:688-701. 122 129. Vasey PA, Kaye SB, Morrison R, 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. Clin Cancer Res 1999;5:83-94. 130. Deng C, Tian HY, Zhang PB, Sun J, Chen XS, Jing XB. Synthesis and characterization of RGD peptide grafted poly(ethylene glycol)-b-poly(L-lactide)-b-poly(L-glutamic acid) triblock copolymer. Biomacromolecules 2006;7:590-6. 131. Farokhzad OC, Cheng JJ, Teply BA, et al. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proceedings of the National Academy of Sciences of the United States of America 2006;103:6315-20. 132. Avgoustakis K. Pegylated poly(lactide) and poly(lactide-co-glycolide) nanoparticles: Preparation, properties and possible applications in drug delivery. Current Drug Delivery 2004;1:321-33. 133. van Vlerken LE, Vyas TK, Amiji MM. Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery. Pharm Res 2007;24:1405-14. 134. Ryan SM, Mantovani G, Wang XX, Haddleton DM, Brayden DJ. Advances in PEGylation of important biotech molecules: delivery aspects. Expert Opin Drug Deliv 2008;5:371-83. 135. Sengupta S, Eavarone D, Capila I, et al. Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system. Nature 2005;436:568-72. 136. Rihova B, Kubackova K. Clinical implications of N-(2-hydroxypropyl)methacrylamide copolymers. Current pharmaceutical biotechnology 2003;4:311-22. 137. Lammers T, Subr V, Peschke P, et al. Image-guided and passively tumour-targeted polymeric nanomedicines for radiochemotherapy. Br J Cancer 2008;99:900-10. 138. Lammers T, Peschke P, Kuhnlein R, et al. Effect of intratumoral injection on the biodistribution and the therapeutic potential of HPMA copolymer-based drug delivery systems. Neoplasia (New York, NY 2006;8:788-95. 139. Duncan R. Polymer conjugates as anticancer nanomedicines. Nature reviews 2006;6:688701. 140. Kim M, Liao J, Dowling ML, et al. TRAIL inactivates the mitotic checkpoint and potentiates death induced by microtubule-targeting agents in human cancer cells. Cancer Research 2008;68:3440-9. 141. Langer R. Drug delivery and targeting. Nature 1998;392:5-10. 142. Liggins RT, Burt HM. Polyether-polyester diblock copolymers for the preparation of paclitaxel loaded polymeric micelle formulations. Advanced Drug Delivery Reviews 2002;54:191-202. 143. De M, Ghosh PS, Rotello VM. Applications of Nanoparticles in Biology. Adv Mater 2008;20:4225-41. 144. Riehemann K, Schneider SW, Luger TA, Godin B, Ferrari M, Fuchs H. NanomedicineChallenge and Perspectives. Angewandte Chemie-International Edition 2009;48:872-97. 145. Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proceedings of the National Academy of Sciences of the United States of America 2008;105:17356-61. 146. Farokhzad OC, Jon SY, Khadelmhosseini A, Tran TNT, LaVan DA, Langer R. Nanopartide-aptamer bioconjugates: A new approach for targeting prostate cancer cells. Cancer Research 2004;64:7668-72. 147. Shojaei F, Ferrara N. Antiangiogenesis to treat cancer and intraocular neovascular disorders. Laboratory Investigation 2007;87:227-30. 148. Hood JD, Bednarski M, Frausto R, et al. Tumor regression by targeted gene delivery to the neovasculature. Science 2002;296:2404-7. 123 149. Dorrell MI, Aguilar E, Scheppke L, Barnett FH, Friedlander M. Combination angiostatic therapy completely inhibits ocular and tumor angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 2007;104:967-72. 150. Poeck H, Besch R, Maihoefer C, et al. '-triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma. Nature Medicine 2008;14:1256-63. 151. Green JJ, Chiu E, Leshchiner ES, Shi J, Langer R, Anderson DG. Electrostatic ligand coatings of nanoparticles enable ligand-specific gene delivery to human primary cells. Nano Letters 2007;7:874-9. 152. Zhang LF, Chan JM, Gu FX, et al. Self-assembled lipid-polymer hybrid nanoparticles: A robust drug delivery platform. Acs Nano 2008;2:1696-702. 153. Sahoo SK, Ma W, Labhasetwar V. Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer. International Journal of Cancer 2004;112:335-40. 154. Tozer GM, Kanthou C, Baguley BC. Disrupting tumour blood vessels. Nature Reviews Cancer 2005;5:423-35. 155. Cheng J, Teply BA, Sherifi I, et al. Formulation of functionalized PLGA-PEG nanoparticles for in vivo targeted drug delivery. Biomaterials 2007;28:869-76. 156. Gu F, Zhang L, Teply BA, et al. Precise engineering of targeted nanoparticles by using self-assembled biointegrated block copolymers. Proceedings of the National Academy of Sciences of the United States of America 2008;105:2586-91. 157. Sengupta S, Eavarone D, Capila I, et al. Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system. Nature 2005;436:568-72. 158. Blagosklonny MV. Antiangiogenic therapy and tumor progression. Cancer Cell 2004;5:13-7. 159. Rofstad EK, Rasmussen H, Galappathi K, Mathiesen B, Nilsen K, Graff BA. Hypoxia promotes lymph node metastasis in human melanoma xenografts by up-regulating the urokinasetype plasminogen activator receptors. Cancer Research 2002;62:1847-53. 160. Kerbel RS. Antiangiogenic therapy: A universal chemosensitization strategy for cancer? Science 2006;312:1171-5. 161. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. New England Journal of Medicine 2004;350:2335-42. 162. Coultas L, Chawengsaksophak K, Rossant J. Endothelial cells and VEGF in vascular development. Nature 2005;438:937-45. 163. Carmeliet P. Angiogenesis in life, disease and medicine. Nature 2005;438:932-6. 164. Holmgren L, Oreilly MS, Folkman J. DORMANCY OF MICROMETASTASES BALANCED PROLIFERATION AND APOPTOSIS IN THE PRESENCE OF ANGIOGENESIS SUPPRESSION. Nature Medicine 1995;1:149-53. 165. Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nature Reviews Cancer 2004;4:423-36. 166. Uno T, Takeda K, Kojima Y, et al. Eradication of established tumors in mice by a combination antibody-based therapy. Nature Medicine 2006;12:693-8. 167. Klement G, Baruchel S, Rak J, et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. Journal of Clinical Investigation 2000;105:R15-R24. 168. Thorpe PE. Vascular targeting agents as cancer therapeutics. Clinical Cancer Research 2004;10:415-27. 169. Devalapally H, Duan ZF, Seiden MV, Amiji MM. Paclitaxel and ceramide coadministration in biodegradable polymeric nanoparticulate delivery system to overcome drug resistance in ovarian cancer. Int J Cancer 2007;121:1830-8. 124 170. Kumar P, Benedict R, Urzua F, Fischbach C, Mooney D, Polverini P. Combination treatment significantly enhances the efficacy of antitumor therapy by preferentially targeting angiogenesis. Laboratory Investigation 2005;85:756-67. 171. Minischetti M, Vacca A, Ribatti D, et al. TNP-470 and recombinant human interferonalpha 2a inhibit angiogenesis synergistically. British Journal of Haematology 2000;109:829-37. 172. Abdollahi A, Griggs DW, Zieher H, et al. Inhibition of alpha(v)beta(3) integrin survival signaling enhances antiangiogenic and antitumor effects of radiotherapy. Clinical Cancer Research 2005;11:6270-9. 173. McCarron PA, Marouf WM, Quinn DJ, et al. Antibody targeting of camptothecin-loaded PLGA nanoparticles to tumor cells. Bioconjugate Chemistry 2008;19:1561-9. 174. McGown AT, Fox BW. STRUCTURAL AND BIOCHEMICAL-COMPARISON OF THE ANTI-MITOTIC AGENTS COLCHICINE, COMBRETASTATIN-A4 AND AMPHETHINILE. Anti-Cancer Drug Design 1989;3:249-54. 175. Kanthou C, Tozer GM. The tumor vascular targeting agent combretastatin A-4-phosphate induces reorganization of the actin cytoskeleton and early membrane blebbing in human endothelial cells. Blood 2002;99:2060-9. 176. Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metal loproteinases. Cancer Cell 2004;6:553-63. 177. Liu Z, Chen K, Davis C, et al. Drug delivery with carbon nanotubes for in vivo cancer treatment. Cancer Research 2008;68:6652-60. 178. Henke E, Perk J, Vider J, et al. Peptide-conjugated antisense oligonucleotides for targeted inhibition of a transcriptional regulator in vivo. Nature Biotechnology 2008;26:91-100. 179. Stoeltzing O, Liu WB, Reinmuth N, et al. Inhibition of integrin alpha(5)beta(1) function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. International Journal of Cancer 2003;104:496-503. 180. Bozec A, Formento P, Lassalle S, Lippens C, Hofman P, Milano G. Dual inhibition of EGFR and VEGFR pathways in combination with irradiation: antitumour supra-additive effects on human head and neck cancer xenografts. British Journal of Cancer 2007;97:65-72. 181. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine 1995;1:27-31. 182. Tran J, Master Z, Yu JL, Rak J, Dumont DJ, Kerbel RS. A role for survivin in chemoresistance of endothelial cells mediated by VEGF. Proceedings of the National Academy of Sciences of the United States of America 2002;99:4349-54. 183. Yang QW, Tian YF, Liu SQ, et al. Thrombospondin-1 peptide ABT-510 combined with valproic acid is an effective antiangiogenesis strategy in neuroblastoma. Cancer Research 2007;67:1716-24. 184. Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nature Reviews Cancer 2002;2:727-39. 185. Deutsch HM, Glinski JA, Hernandez M, et al. Synthesis of congeners and prodrugs.3. water-soluble prodrugs of Taxol with potent antitumor activity. Journal of Medicinal Chemistry 1989;32:788-92. 186. Woodrow KA, Cu Y, Booth CJ, Saucier-Sawyer JK, Wood MJ, Saltzman WM. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nature Materials 2009;8:526-33. 187. Satchi-Fainaro R, Puder M, Davies JW, et al. Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470. Nature Medicine 2004;10:255-61. 188. Chaplin DJ, Pettit GR, Hill SA. Anti-vascular approaches to solid tumour therapy: Evaluation of combretastatin A4 phosphate. Anticancer Research 1999;19:189-95. 189. van Vlerken LE, Duan Z, Little SR, Seiden MV, Amiji MM. Biodistribution and pharmacokinetic analysis of paclitaxel and ceramide administered in multifunctional polymer125 blend nanoparticles in drug resistant breast cancer model. Molecular Pharmaceutics 2008;5:51626. 190. Barberaguillem E, Smith I, Weiss L. CANCER-CELL TRAFFIC IN THE LIVER .1. GROWTH-KINETICS OF CANCER-CELLS AFTER PORTAL-VEIN DELIVERY. International Journal of Cancer 1992;52:974-7. 191. Pasquier E, Carre M, Pourroy B, et al. Antiangiogenic activity of paclitaxel is associated with its cytostatic effect, mediated by the initiation but not completion of a mitochondrial apoptotic signaling pathway. Mol Cancer Ther 2004;3:1301-10. 192. Benny O, Fainaru O, Adini A, et al. An orally delivered small-molecule formulation with antiangiogenic and anticancer activity. Nature Biotechnology 2008;26:799-807. 193. Dobrovolskaia MA, Germolec DR, Weaver JL. Evaluation of nanoparticle immunotoxicity. Nature Nanotechnology 2009;4:411-4. 194. Yoo HS, Oh JE, Lee KH, Park TG. Biodegradable nanoparticles containing doxorubicinPLGA conjugate for sustained release. Pharmaceutical Research 1999;16:1114-8. 195. Tong R, Cheng JJ. Ring-Opening Polymerization-Mediated Controlled Formulation of Polylactide-Drug Nanoparticles. Journal of the American Chemical Society 2009;131:4744-54. 126 [...]... Pharmaceutical Research (In Press) 6 Wang Z., Ho PC A nanocapsular combinatorial sequential drug delivery system for antiangiogenesis and anticancer activities Biomaterials 2010; 31(27), 7115-7123 7 Wang Z., Ho PC Core-shell nanocapsule targeting to the tumor vasculature with sequential drug delivery for antivasculature and anticancer activities Small 2010; 6(22), 2576-2583 8 Wang Z., Lee T.Y., Ho PC cRGD-dextran-oleate... KP CdSe/AsS Core-Shell Quantum Dots: Preparation and Two-Photon Fluorescence J Am Chem Soc 2009; 131(32), 1130011301 4 Wang Z., Chui WK., Ho PC Integrin targeted drug and gene delivery Expert opinion on drug delivery 2010; 7(2), 159-171 5 Wang Z., Chui WK Ho PC Nanoparticulate delivery system targeted to tumor neovasculature for combined anticancer and antiangiogenesis therapy Pharmaceutical Research... these characteristics make integrin a good targeting ligand for both tumor and antiangiogenesis targeting therapy In recent years, Jain and co-workers have proposed that certain antiangiogenetic agents can transiently normalize the abnormal structure and function of the tumor vasculature, and make it more efficient for oxygen, nutrients and drug delivery (67) Thus, combination of vascular 15 targeting... survival time and much less, if not at all, metastasis spots in animal liver when compared with other controls In conclusion, our results supported the hypothesis that the nanoparticulated combinatorial drug delivery system for simultaneous antiangiogenesis and anticancer activities could improve the therapeutic outcome The nanoparticle formulations described in this thesis were all simple, robust and easy... occurrence Table 1-2 lists some of antiangiogenesis inhibitors in different phases of clinical trials 1.3 Drug delivery system for vascular targeted antiangiogenesis or antitumor therapy 1.3.1 Conjugation of drug with ligand for vascular targeting Integrins are important extracellular molecules that regulate angiogenesis process They are transmembrane glycoproteins, and can be internalized by cells upon... nanoparticle for targeted chemotherapeutic delivery of paclitaxel (Submitted for review) 9 Wang Z., Liew G F., Ho PC Double targeting cRGD-HA-DSPE self-assembled PLGA core-shell nanoparticle for the delivery of paclitaxel to breast cancer cells (Submitted for review) XI List of Tables Table 1-1 List of the integrin subunit combinations, their distribution, and RGD recognition 13 1-2 Selected antiangiogenesis. .. according to their environment and interactions with adjacent cells (29) Each step is essential to the formation of tumor associated neovasculature and represents a potential site of therapeutic manipulation 8 Tumor associated angiogenesis often develops in a hazard and poorly regulated manner The resultant vessels tend to form right-angle turns and corkscrew spirals, and fail to diminish progressively... proteins Bcl-2 and Bcl-XL and the proapoptotic protein Bax (46) Another widely accepted method for antiangiogenesis therapy is the interference of angiogenetic growth factor such as VEGF, PDGF or their receptors Typically, VEGF plays a vital role in the tumor development and angiogenesis Hence, the blockage of VEGF function could effectively inhibit the angiogenesis and subsequent tumor growth and metastasis... of VEGF receptor cellular signaling (50), etc Intervention with endothelial cell adhesion and migration: Because interaction between endothelial cell and extracellular matrix is highly involved in the angiogenesis progress for endothelial cell adhesion and migration, many efforts have been paid on this step for antiangiogenesis purpose Interferon (IFN), the first identified endogenous angiogenesis inhibitor,... remodeling and morphogenesis These enzymes are capable of degrading all components of the extracellular matrix (64) Increased activity of these enzymes has been observed in tumor formation, and therefore inhibitors of MMPs represent an attractive approach to treat cancer Batimastat, marimastat, and prinomastat are all potent broad14 spectrum MMPs inhibitors, and can prevent or reduce growth, angiogenesis and . DRUG DELIVERY SYSTEMS FOR ANTIANGIOGENESIS AND ANTICANCER ACTIVITIES WANG ZHE A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY. nanoparticulated combinatorial drug delivery system for simultaneous antiangiogenesis and anticancer activities could improve the therapeutic outcome. The nanoparticle formulations described in. 1.3.1 Conjugation of drug with ligand for vascular targeting 15 1.3.2 Vascular targeted nanoparticle drug delivery 19 1.4 Conclusion 22 Chapter 2 Targeted nanoparticulate drug delivery system to

Ngày đăng: 11/09/2015, 09:58

Từ khóa liên quan

Mục lục

  • cover

  • ACKNOWLEDGEMENTS

  • Main contentThesis Dec 1.2010-wz

    • Chapter 1

    • Introduction

      • 1.1 Cancer

        • 1.1.1 Introduction

        • 1.1.2 Metastasis of cancer

        • 1.1.3 Angiogenesis of cancer

        • 1.1.4 Relationship of angiogenesis with tumor growth and metastasis

        • 1.2 Antiangiogenesis therapy

          • 1.2.1 Antiangiogenesis inhibitors

          • 1.3 Drug delivery system for vascular targeted antiangiogenesis or antitumor therapy

            • 1.3.1 Conjugation of drug with ligand for vascular targeting

            • 1.3.2 Vascular targeted nanoparticle drug delivery

            • 1.4 Conclusion

            • Chapter 2

            • Targeted nanoparticulate drug delivery system to malignant cancer cells

              • 2.1 Introduction

              • 2.2 Materials and methods

                • 2.2.1 Materials

                • 2.2.2 Conjugation of doxorubicin to PLGA

                • 2.2.3 Preparation of PLGA-doxorubicin (PLGA-Doxo) nanoparticles

                • 2.2.4 Synthesis of Doxo-PLGA-PEG-cRGD nanoparticles

                • 2.2.5 Characterization of nanoparticles

                • 2.2.6 Cytotoxicity assay

                • 2.2.7 Cell uptake and binding affinity assays

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan