Use of upconversion fluorescence nanoparticles in biomedical applications

167 345 0
Use of upconversion fluorescence nanoparticles in biomedical applications

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

USE OF UPCONVERSION FLUORESCENCE NANOPARTICLES IN BIOMEDICAL APPLICATIONS DEV KUMAR CHATTERJEE (M.B.B.S., M.M.S.T) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DIVISION OF BIOENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2007 _______________________________________________________________________________ PREFACE This thesis is hereby submitted for the degree of Doctor of Philosophy in the Division of Bioengineering at the Faculty of Engineering, National University of Singapore. This thesis, either in part or whole, has never been submitted for any other degree or equivalent to another university or institution. This thesis contains all original work, unless specifically mentioned and referenced to other works. Parts of this thesis has been published or presented in: Peer reviewed journal publications: Chatterjee, D.K., Fong L.S., Zhang Y., Nanoparticles in photodynamic therapy: an emerging paradigm. Advanced Drug Delivery Reviews (Invited article, under review) Chatterjee, D.K., Zhang, Y. Upconverting Nanoparticles as Nano-Transducers for Photodynamic Therapy in Cancer Cells, Nanomedicine Vol. 3, No. (2008) 73-82. Chatterjee, D.K., Zhang, Y. Upconversion fluorescence imaging of cells and small animals using lanthanide doped nanocrystals, Biomaterials Volume 29, Issue 7, (2008) 937-943 Wang, F., Chatterjee, D.K., Li, Z.Q., Zhang, Y., Fan, X.P., Wang, M.Q. Synthesis of polyethylenimine/NaYF4 nanoparticles with upconversion fluorescence, Nanotechnology, vol. 17, No 23 (14 December 2006) 5786-5791 ii _______________________________________________________________________________ Review book chapters Chatterjee D.K. and Zhang, Y. (2007). Lanthanide doped Upconverting Nanoparticles for biomedical applications. Doped Nanomaterial and Nanodevices. Wei Chen. American Scientific Publishers (in press). Chatterjee, D.K. and Y. Zhang (2007). Nanoparticles in Immunotherapy Against Cancer. Cancer Nanotechnology – Nanomaterials for Cancer Diagnosis and Therapy. H. S. Nalwa and T. Webster. Valencia, American Scientific Publishers. 317 - 332 Zhang, Y. and Chatterjee D.K. (2006). Liposomes, dendrimers and other polymeric nanoparticles for targeted delivery of anticancer agents - A comparative study. Nanomaterials for Cancer Therapy. C. S. S. R. Kumar. Weinheim, Wiley-VCH Verlag GmbH & Co, KGaA. 6: 338 - 370. Conference abstracts: Chatterjee DK and Zhang Y, Upconverting Nanoparticles as Nano-Transducers for Photodynamic Therapy in Cancer Cells. (NSTI Nanotechnology Conference and Trade Show, June 1-5, 2008, in Boston, Massachusetts, U.S.A) Chatterjee DK and Zhang Y, Upconverting Nanoparticles for in vitro and in vivo imaging. 2008 (NSTI Nanotechnology Conference and Trade Show, June 1-5, 2008, in Boston, Massachusetts, U.S.A) iii _______________________________________________________________________________ Chatterjee D.K. and Zhang Y., Up-converting Nanoparticles: Novel Soluble Probes for Imaging of Live Cancer Cells and Tissues. 2007 Spring Proceedings; Volume 1019E 1019-FF08-11 (2007). Moscone West: San Francisco Marriott, San Francisco, CA, USA (2007 MRS Spring Meeting, April 9—13, 2007) Zhang Y and Chatterjee DK, Multi-functional nanoparticles for cancer therapy. Abstract Book of International Symposium on Nanotechnology in Environmental Protection and Pollution (2006): 31. Hong Kong: The Hong Kong University of Science & Technology .(International Symposium on Nanotechnology in Environmental Protection and Pollution, 18 -21 Jun 2006, The Hong Kong University of Science & Technology, Hong Kong, China) Chatterjee, D.K., Zhang, Y. Multi-functional nanoparticles for cancer therapy, Science and Technology of Advanced Materials, vol (2007) 131-133 Chatterjee DK and Zhang Y, Evaluation of the biocompatibility of the bi-functional nanoparticles. Proceedings of The 12th International Conference on Biomedical Engineering (2005). Singapore: IFMBE. (The 12th International Conference on Biomedical Engineering (ICBME 2005), - 10 Dec 2005, Singapore) Chatterjee DK and Zhang Y, Synthesis and Characterization of Bi-functional Nanoparticles for Cancer Immunotherapy. Proceedings of The 12th International Conference on Biomedical Engineering (2005). Singapore: IFMBE. (The 12th International Conference on Biomedical Engineering (ICBME 2005), - 10 Dec 2005, Singapore) iv _______________________________________________________________________________ ACKNOWLEDGEMENTS I would like to acknowledge the contributions of my guide A/Prof Zhang Yong for his constant encouragement, guidance and advice without which none of this would have been possible. I have also been supported during this long effort by my colleagues who have taught me procedures or helped with the synthesis of the nanoparticles. The help from Initha Appavoo, Dr Li Zhengquan (nanoparticles) and Dr Rufaihah (animal experiments) deserve a special mention. A special note of thanks to those undergraduates – primarily Lim Sock Yong, Xiuli and Eliza – who have put in long hard hours and challenged me with their constant queries. All have contributed to make this journey not only a learning one but also an enjoyable one. I would also like to acknowledge the research grant from the National University of Singapore for the essential financial support. Finally, my thanks to my family - and especially my wife Deyali - whose constant love and support helped me through the toughest times. Dev Kumar Chatterjee April, 2008 v _______________________________________________________________________________ TABLE OF CONTENTS PREFACE . II ACKNOWLEDGEMENTS V TABLE OF CONTENTS . VI SUMMARY IX LIST OF TABLES . XII LIST OF FIGURES XIII ABBREVIATIONS . XVI CHAPTER LITERATURE REVIEW & RESEARCH PROGRAM . 1.1 Definition and scope . 1.2 Nanoparticles for disease diagnostics 1.2.1 Molecular targeting using nanoparticles . 1.2.2 Fluorescent nanoparticles as imaging probes 1.3 Nanoparticles in therapeutic applications . 16 1.3.1 General principles 16 1.3.2 Nanoparticles for photodynamic therapy of cancer . 19 1.4 Upconversion nanoparticles . 26 1.4.1 Principle of upconversion 26 1.4.2 Upconversion nanoparticles: definition and materials . 29 1.4.3 Surface modifications of upconverting nanoparticles . 32 1.5 Thesis overview 36 CHAPTER SYNTHESIS & CHARACTERIZATION OF UPCONVERSION NANOPARTICLES . 40 2.1 Introduction 41 2.2 Materials and Methods 44 2.2.1 Reagents 44 2.2.2 Synthesis of PEI/NaYF4 nanoparticles . 45 2.2.3 Physical characterization of the nanoparticles 45 2.2.4 Optical characterization . 48 vi _______________________________________________________________________________ 2.2.5 Cell biocompatibility test . 52 2.3 Results and Discussion . 55 2.3.1 Physical characterization of the nanoparticles 55 2.3.2 Optical characterization of the nanoparticles 61 2.3.3 Cell viability test 73 2.4 Conclusion 78 CHAPTER IMAGING OF CANCER CELLS USING UPCONVERSION NANOPARTICLES . 79 3.1 Introduction 80 3.2 Materials and Methods 81 3.2.1 Materials 81 3.2.2 Attachment of targeting ligand on upconversion nanoparticles . 81 3.2.3 Size measurement with TEM . 82 3.2.4 Surface charge measurement 82 3.2.5 Detection of aggregates in solution 83 3.2.6 Detection of folic acid on the nanoparticles 83 3.2.7 Incubation of nanoparticles with cancer cells . 84 3.2.8 Confocal imaging . 84 3.2.9 Efficiency and specificity of targeting of nanoparticles to cancer cells 87 3.3 Results . 88 3.3.1 TEM of FA-PEI/NaYF4 . 88 3.3.2 Confirmation of folic acid binding on nanoparticles by FTIR . 89 3.3.3 Alteration of zeta-potential due to folic acid attachment . 90 3.3.4 Detection of aggregates by size distribution . 90 3.3.5 Imaging of cancer cells 91 3.3.6 Effect of incubation period on uptake of nanoparticles . 94 3.4 Conclusion 99 CHAPTER UPCONVERSION NANOPARTICLES FOR IN VIVO IMAGING 100 4.1 Introduction 101 4.2 Materials and Methods 104 4.2.1 Materials 104 4.2.2 Imaging of upconversion nanoparticles within rat skin . 105 4.2.3 Comparison of upconversion nanoparticles with QDs for in vivo imaging 105 4.2.4 Imaging of upconversion nanoparticles in other rat tissues . 106 4.2.5 In vivo microscopy using upconversion nanoparticles 106 4.3 Results and Discussion . 109 4.3.1 Imaging of subcutaneously injected upconversion nanoparticles vii _______________________________________________________________________________ . 109 4.3.2 Comparative imaging of subcutaneous injection of nanoparticles 111 4.3.3 Imaging of injected nanoparticles in other tissues . 112 4.3.4 In vivo cell imaging . 113 4.4. Conclusion . 116 CHAPTER UPCONVERSION NANOPARTICLES IN PHOTODYNAMIC THERAPY OF CANCER .117 5.1 Introduction 118 5.2 Materials and Methods 120 5.2.1 Materials 120 5.2.2 Preparation of ZnPC standard curve by spectrophotometry 121 5.2.3 Attaching ZnPC to FA-PEI/NaYF4:Yb,Er nanoparticles 121 5.2.4 Detection of ZnPC on the surface of the nanoparticles . 123 5.2.5 Determination of singlet oxygen production . 123 5.2.6 Targeted binding to human cancer cells 124 5.2.7 Photoexposure of cells . 124 5.2.8 MTT assay to check effectiveness of PDT 124 5.3 Results and Discussion . 126 5.3.1 Standard curve for ZnPC 126 5.3.2. Encapsulation efficiency . 126 5.3.3 FTIR for presence of ZnPC 127 5.3.4 Spectroscopy to determine emission-excitation overlap 128 5.3.5 Singlet oxygen production by ADPA molecular probe 129 5.3.6 Targeted uptake of ZnPC-UCN by cancer cells 131 5.3.7 Effectiveness of PDT using ZnPC-UCN . 132 5.3.8 Effect of nanoparticle concentration . 134 5.4 Conclusion 135 CHAPTER CONCLUSION AND FUTURE WORK 137 REFERENCES 142 viii _______________________________________________________________________________ SUMMARY Nanoparticles are spherical aggregates less than 100nm in diameter containing a few hundreds to thousands of atoms. Fluorescent nanoparticles excited by near infrared (NIR) are advantageous because NIR gives rise to minimal autofluorescence which results in very high signal-to-background ratios; cells and tissue destruction is low because NIR is harmless to biomolecules in low doses; and nanoparticles can be imaged from inside tissues because of deep penetration of NIR radiation. This thesis explores the characterization and biomedical applications of a new variety of NIR excited fluorescent nanoparticles, PEI/NaYF44:Er,Yb, developed at the Cellular and Molecular Bioengineering Laboratory, with a focus on cancer. PEI/NaYF4 upconversion nanoparticles co-doped with Er and Yb were demonstrated to be 60 nm spherical particles of uniform shape and size, positive surface charge and stably soluble in de-ionized water. When excited with 980nm laser these emitted light with sharp peaks in the red and green region of the visible spectrum. This emission was strongly photostable and immune to storage over weeks, although incubation in serum at physiological temperatures slowly degraded the signal, probably by protein deposition. The particles were biocompatible with two different human cell lines to moderately high concentrations and for reasonable periods of incubation. The upconverting nanoparticles (UCN) were conjugated to a cell-specific ligand and used for targeted imaging of live human cancer cells in vitro. These showed strong signal-to-background ratios and high sensitivity of detection. Non-targeted tagging of cells using PEI polymer as a positively charged coupler was also demonstrated. All imaging experiments showed signal stability and absence of cell damage as a result of ix _______________________________________________________________________________ prolonged laser exposure. The ability to image these nanoparticles inside animals was demonstrated by injecting into various tissues of live, anaesthetized rats and exciting the injection site with NIR laser. Fluorescence from injected nanoparticles was recorded at injection depths of a few millimeters to nearly cm, the depth depending on the type of tissue injected, the dose of nanoparticles and the effective control of ambient light which contributes to background. Human cancer cells, non-specifically tagged with upconversion nanoparticles, were injected subcutaneously in live anaesthetized mice and the cells imaged by real-time in vivo confocal microscopy. Photodynamic therapy (PDT) is a therapeutic option for cancer that relies on the interaction of light and photosensitizer drugs to kill targeted cells. Acceptance of PDT has been limited by, among other factors, fear of high cost of setup and inability to easily reach deeper seated tumors. We demonstrated a nanoparticle-based approach to address these problems. UCN were functionalized with zinc phthalocyanine (ZnPC) photosensitizer for simultaneous imaging and photodynamic therapy. The nanoparticles act as ‘nano-transducers’ to convert deeply tissue penetrating NIR excitation to emission frequencies suitable to activate the photosensitizer to release reactive oxygen species to kill cancer cells. The effectiveness of the modified nanoparticles for this purpose was demonstrated in vitro with concurrent imaging. Both the imaging and photodynamic therapy of cancer cells using PEI/NaYF44:Er,Yb nanoparticles were described, to the best of my knowledge, for the first time, although several preliminary results using large phosphor ‘nanoparticles’ in excess of 100nm in diameter can be found in the literature. These results lead the slow emergence of the phosphor nanoparticles as a valuable fluorescent label for biomedical applications, set x _______________________________________________________________________________ had been demonstrated that coating with folic acid promoted selectivity of binding to cancer cells over normal cells (e.g. fibroblasts). Hence, it can be reasonably assumed that the FA-ZnPC-UCN demonstrated here will also show selectivity towards cancer cells. In conclusion, we have described a nanodevice for cancer PDT where upconverting nanoparticles convert deeply penetrating NIR light to visible emissions, which are in turn used by the attached photosensitizer to convert molecular oxygen to toxic singlet species. It can be appreciated that this is essentially a platform technology with the scope to substitute ZnPC with other photosensitizers which absorb in the 540nm or 650nm range. For example, tin etiopurpurin (SnET2) requires 660nm while tetra(mhydroxyphenyl)chlorine (mTHPC, Foscan) is excited at 652nm. Further studies into effectiveness of this therapy in animals are being explored. 136 Chapter CONCLUSION AND FUTURE WORK _______________________________________________________________________________ Fluorescent labels that are excited by the near infrared (NIR) radiation give rise to minimal autofluorescence because of lack of efficient endogenous absorbers in the NIR spectral range (Bremer et al., 2001; Konig, 2000; Larson et al., 2003) and the absence of upconversion among most biomolecules. This results in very high signal to background ratios. Photo-damage to cells and tissues is also greatly reduced because NIR is generally harmless to biomolecules in low doses. Penetration depths are increased several orders of magnitude (from a few mm for UV to up to as deep as 15 cm in fibrofatty tissues like breast tissues for NIR). (Auzel, 2004; Soukka et al., 2005). The rare earth elements used in synthesis of upconversion nanoparticles have a lower toxicity than semiconductor elements of quantum dots. These chemically stable particles not bleach and allow permanent excitation with simultaneous signal integration (Corstjens et al., 2005). Since upconversion occurs within the host crystal, optical properties are unaffected by the environment (e.g., chemicals, pH and temperature). Consequently, use of these particles is unaffected by the nature of the analyte in assays (such as whole blood, plasma, urine, sputum, or tissue homogenates) or by the microenvironment in cellular and subcellular imaging. UCNs typically have narrower absorption and line emission spectra, as compared to the downconverting fluorophores. The emission of upconverting particles consist of sharp lines characteristic of atomic transitions in a well-ordered matrix. To exploit these advantages, upconverting nanoparticles, or UCN, need to be developed and explored. For cancer cell targeting, folic acid was covalently conjugated to the PEI/NaYF4 nanoparticles. The method of cross-linking FA to PEI involves a simple condensation reaction and the substitution of folic acid by other biomolecules such as antibodies or peptides can be carried out without significant alteration of the synthesis protocol. In 138 _______________________________________________________________________________ these experiments the human colonic adenocarcinoma cell line HT29 and the ovarian cancer cell line OVCAR3 were used because they possess high amounts of folate receptors on their surface. Live cells were imaged and the images showed the attachment of the upconversion nanoparticles onto the surface of the cells. While the binding of the nanoparticles to the surface of the cells is rapid and extensive fluorescence can be detected within an hour, uptake inside cells through the internalization of the bound receptor-nanoparticle complex is a much slower process and can usually be detected in significant amount only after 48 hours of incubation. However, using NIR to image cells has a disadvantage too. Since the maximum resolution achievable with microscope varies inversely with the wavelength of light used, using NIR imposes a theoretical limit on the quality of the achieved images. This can be seen in the fluorescent images obtained where small bright clusters of the UCN appear to be diffuse masses of nanoparticles. However, in the tradeoff between resolution and sensitivity, NIR presents reasonably high values of both. Uncoated nanoparticles are covered with a surface layer of PEI which is a highly positively charged molecule with a large number of amino groups. It is to be expected that these nanoparticles will be involved in formation of hydrogen bonds with negatively charged molecules on the surface of the cells and also on the free surface of the well plate. These nanoparticles thus show a moderately high non-specific binding to cell surfaces after incubation. Coating with folic acid, however, significantly lowers the amount of non-specific binding and spurious background luminescence can be avoided with thorough washing. 139 _______________________________________________________________________________ However, much can be done to maximize the advantageous use of these nanoparticles in non-invasive live cell imaging. The simplicity of a one-pot synthesis and a universal binding mechanism for any carboxylate group containing ligand lends itself to ease of exploring variations of this theme. Substitutions with antibodies or peptides for more specific targeting can be a step ahead. The reduction of fluorescence degradation in biofluids can be utilized for long term imaging of cellular processes in vitro and in vivo. Addition of other molecules like drugs and biologic response modifiers to confer multi-functional nanodevices is an attractive field of exploration. The animal experiments were carried out using an imaging system equipped with a 980nm laser and simple CCD cameras. Much can be done to improve this setup including mobile imaging platforms to record whole body scans of the animals. Better black-box arrangements can make possible significantly improved signal-to-noise ratios. Laser power can be optimized for maximal luminescence with minimal tissue damage although tissues not have a strong absorption at the wavelength of 980nm. The scope and possible benefits of continuous imaging subcutaneous and muscular tissues in real time is immense and needs to be thoroughly explored. To the best of our knowledge, this represents the first study of imaging of live cells and deep tissues in animal using upconversion fluorescent nanoparticles. Photodynamic therapy of superficial and slightly deeper tumors is an exciting application for these nanoparticles. The next step, the testing of these nanoparticles in small animal model of tumor, has already started in our laboratory. We have started experiments on a bladder and skin tumor model for mice. It is an exciting area of research with potential to significantly affect biomedical diagnostics and therapeutics. 140 _______________________________________________________________________________ In conclusion, UCN present an additional tool to the biologist working with fluorescent nanoparticles. However, in no way these supplant existing fluorescent nanoparticles that have become popular with many biologists and researchers. One of the many reasons for that is the existing fluorescent nanoparticles are very much brighter than the UCN. For nearly all biological experiments, the most important properties of a fluorescent label are brightness and stability. Quantum dots have both qualities. Another advantage of commercially available fluorescent nanoparticles is the much smaller size than UCN. QDs are usually of the order of a few nanometers, usually less than 10nm. This size being similar to that of the average protein molecule, QDs can be used in most biological experiments without hindrance. In contrast, UCN are several times in diameter to QDs and often less bright. While multicolor UCN have been demonstrated, QDs present a larger range of colors mainly because unlike UCN they can tuned by their size and not on the type of impurity. Having said all that, it is also to be noted that UCN are progressing towards being a mature technology, with more uniform particles of smaller diameter being reported every year. The main roles of UCN would not be replace QDs, but to present an alternative fluorescent label that has some specialized properties that can be profitably exploited for specific needs. This thesis aims to demonstrate a few of these. 141 _______________________________________________________________________________ REFERENCES Auzel F. 2004.Upconversion and anti-stokes processes with f and d ions in solids. Chemical Reviews;104(1):139-73. Auzel F. 2004.Upconversion and anti-Stokes processes with f and d ions in solids. Chem Rev;104(1):139-73. Babacan S, Pivarnik P, Letcher S, Rand AG. 2000.Evaluation of antibody immobilization methods for piezoelectric biosensor application. Biosensors & Bioelectronics;15(11-12):615-21. Bahulekar R, Ayyangar NR, Ponrathnam S. 1991.Polyethyleneimine In Immobilization Of Biocatalysts. Enzyme And Microbial Technology;13(11):858-68. Baker A, Saltik M, Lehrmann H, Killisch I, Mautner V, Lamm G, Christofori G, Cotten M. 1997.Polyethylenimine (PEI) is a simple, inexpensive and effective reagent for condensing and linking plasmid DNA to adenovirus for gene delivery. Gene Therapy;4(8):773-82. Bargmann CI, Hung MC, Weinberg RA. 1986.The neu oncogene encodes an epidermal growth factor receptor-related protein. Nature;319(6050):226-30. Bazzi R, Flores MA, Louis C, Lebbou K, Zhang W, Dujardin C, Roux S, Mercier B, Ledoux G, Bernstein E, Perriat P, Tillement O. 2004.Synthesis and properties of europium-based phosphors on the nanometer scale: Eu2O3, Gd2O3:Eu, and Y2O3:Eu. J Colloid Interface Sci;273(1):191-7. Birrenbach G, Speiser PP. 1976.Polymerized micelles and their use as adjuvants in immunology. J Pharm Sci;65(12):1763-6. Borg A, Tandon AK, Sigurdsson H, Clark GM, Ferno M, Fuqua SA, Killander D, McGuire WL. 1990.HER-2/neu amplification predicts poor survival in nodepositive breast cancer. Cancer Res;50(14):4332-7. Boyer JC, Vetrone F, Cuccia LA, Capobianco JA. 2006.Synthesis of colloidal upconverting NaYF44 nanocrystals doped with Er3+, Yb3+ and Tm3+, Yb3+ via thermal decomposition of lanthanide trifluoroacetate precursors. J Am Chem Soc;128(23):7444-5. Bremer C, Tung CH, Weissleder R. 2001.In vivo molecular target assessment of matrix metalloproteinase inhibition. Nature Medicine;7(6):743-8. Brigger I, Dubernet C, Couvreur P. 2002.Nanoparticles in cancer therapy and diagnosis. Adv Drug Deliv Rev;54(5):631-51. 142 _______________________________________________________________________________ Browning M. The cancer cell and the immune system. New York: John Wiley & Sons, 1995:99-122. Cai W, Shin DW, Chen K, Gheysens O, Cao Q, Wang SX, Gambhir SS, Chen X. 2006.Peptide-labeled near-infrared quantum dots for imaging tumor vasculature in living subjects. Nano Lett;6(4):669-76. Campbell IG, Jones TA, Foulkes WD, Trowsdale J. 1991.Folate-binding protein is a marker for ovarian cancer. Cancer Res;51(19):5329-38. Carrel S, Zografos L, Schreyer M, Rimoldi D. 1993.Expression of CALLA/CD10 on human melanoma cells. Melanoma Res;3(5):319-23. Chatterjee DK, Zhang Y. 2007.Upconversion fluorescence imaging of cells and small animals using lanthanide doped nanocrystals. Biomaterials;In press( Chawla JS, Amiji MM. 2002.Biodegradable poly(epsilon -caprolactone) nanoparticles for tumor-targeted delivery of tamoxifen. Int J Pharm;249(1-2):127-38. Colcher D, Milenic DE, Ferroni P, Roselli M, Schlom J. 1991.In vivo and in vitro clinical applications of monoclonal antibodies against TAG-72. Int J Rad Appl Instrum B;18(4):395-401. Coney LR, Tomassetti A, Carayannopoulos L, Frasca V, Kamen BA, Colnaghi MI, Zurawski VR, Jr. 1991.Cloning of a tumor-associated antigen: MOv18 and MOv19 antibodies recognize a folate-binding protein. Cancer Res;51(22):6125-32. Corstjens P, Zuiderwijk M, Brink A, Li S, Feindt H, Niedbala RS, Tanke H. 2001.Use of up-converting phosphor reporters in lateral-flow assays to detect specific nucleic acid sequences: a rapid, sensitive DNA test to identify human papillomavirus type 16 infection. Clin Chem;47(10):1885-93. Corstjens PL, Li S, Zuiderwijk M, Kardos K, Abrams WR, Niedbala RS, Tanke HJ. 2005.Infrared up-converting phosphors for bioassays. IEE Proc Nanobiotechnol;152(2):64-72. Corstjens PL, Zuiderwijk M, Nilsson M, Feindt H, Sam Niedbala R, Tanke HJ. 2003.Lateral-flow and up-converting phosphor reporters to detect singlestranded nucleic acids in a sandwich-hybridization assay. Anal Biochem;312(2):191-200. Courvreur P GL, Lenaerts V, Brasseur F, Guiot P, Biornacki A. Biodegradable polymeric nanoparticles as drug carrier for antitumor agents.: Boca Raton, 1986:1. 27-93. Coussens L, Yang-Feng TL, Liao YC, Chen E, Gray A, McGrath J, Seeburg PH, Libermann TA, Schlessinger J, Francke U, et al. 1985.Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene. Science;230(4730):1132-9. 143 _______________________________________________________________________________ de Nonancourt-Didion M, Gueant JL, Adjalla C, Chery C, Hatier R, Namour F. 2001.Overexpression of folate binding protein alpha is one of the mechanism explaining the adaptation of HT29 cells to high concentration of methotrexate. Cancer Lett;171(2):139-45. Duncan R. 2003.The dawning era of polymer therapeutics. Nat Rev Drug Discov;2(5):347-60. Feng W, Dev KC, Zhengquan L, Yong Z, Xianping F, Minquan W. 2006.Synthesis of polyethylenimine/NaYF44 nanoparticles with upconversion fluorescence. Nanotechnology;17(23):5786. Ferrari M. 2005.Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer;5(3):161-71. Florence AT, Hussain N. 2001.Transcytosis of nanoparticle and dendrimer delivery systems: evolving vistas. Adv Drug Deliv Rev;50 Suppl 1(S69-89. Fortina P, Kricka LJ, Surrey S, Grodzinski P. 2005.Nanobiotechnology: the promise and reality of new approaches to molecular recognition. Trends Biotechnol;23(4):168-73. Frangioni JV. 2003.In vivo near-infrared fluorescence imaging. Curr Opin Chem Biol;7(5):626-34. Franken PA, Hill AE, Peters CW, Weinreich G. 1961.Generation of Optical Harmonics. Physical Review Letters;7(4):118. Fu A, Gu W, Larabell C, Alivisatos AP. 2005.Semiconductor nanocrystals for biological imaging. Curr Opin Neurobiol;15(5):568-75. Garin-Chesa P, Campbell I, Saigo PE, Lewis JL, Jr., Old LJ, Rettig WJ. 1993.Trophoblast and ovarian cancer antigen LK26. Sensitivity and specificity in immunopathology and molecular identification as a folate-binding protein. Am J Pathol;142(2):557-67. Godbey WT, Wu KK, Mikos AG. 1999.Poly(ethylenimine) and its role in gene delivery. J Control Release;60(2-3):149-60. Hakomori S. Tumor-associated carbohydrate markers: chemical and physical basis and cell biological implications. Totowa, NJ: The Human Press, 1992:207-32. Hakomori S. 1996.Tumor malignancy defined by aberrant glycosylation and sphingo(glyco)lipid metabolism. Cancer Res;56(23):5309-18. Hampl J, Hall M, Mufti NA, Yao YM, MacQueen DB, Wright WH, Cooper DE. 2001.Upconverting phosphor reporters in immunochromatographic assays. Anal Biochem;288(2):176-87. Hattori Y, Maitani Y. 2004.Enhanced in vitro DNA transfection efficiency by novel folate-linked nanoparticles in human prostate cancer and oral cancer. J Control Release;97(1):173-83. 144 _______________________________________________________________________________ Heer S, Kompe K, Gudel HU, Haase M. 2004.Highly efficient multicolour upconversion emission in transparent colloids of lanthanide-doped NaYF44 nanocrystals. Advanced Materials;16(23-24):2102-5. Heer S, Lehmann O, Haase M, Gudel HU. 2003.Blue, green, and red upconversion emission from lanthanide-doped LuPO4 and YbPO4 nanocrystals in a transparent colloidal solution. Angewandte Chemie-International Edition;42(27):3179-82. Helander IM, Alakomi HL, Latva-Kala K, Koski P. 1997.Polyethyleneimine is an effective permeabilizer of gram-negative bacteria. Microbiology (Reading, England);143 (Pt 10)(3193. Hirabayashi Y, Hamaoka A, Matsumoto M, Matsubara T, Tagawa M, Wakabayashi S, Taniguchi M. 1985.Syngeneic monoclonal antibody against melanoma antigen with interspecies cross-reactivity recognizes GM3, a prominent ganglioside of B16 melanoma. J Biol Chem;260(24):13328-33. Holm J, Hansen SI, Hoier-Madsen M. 1991.High-affinity folate binding in human liver membranes. Biosci Rep;11(3):139-45. Jain KK. 2005.Nanotechnology-based Drug Delivery for Cancer. Technol Cancer Res Treat;4(4):407-16. Jaiswal JK, Mattoussi H, Mauro JM, Simon SM. 2003.Long-term multiple color imaging of live cells using quantum dot bioconjugates. Nat Biotechnol;21(1):47-51. Kawashima Y. 2001.Panoparticulate systems for improved drug delivery. Adv Drug Deliv Rev;47(1):1-2. Keer HN, Kozlowski JM, Tsai YC, Lee C, McEwan RN, Grayhack JT. 1990.Elevated transferrin receptor content in human prostate cancer cell lines assessed in vitro and in vivo. J Urol;143(2):381-5. Kim S, Lim YT, Soltesz EG, De Grand AM, Lee J, Nakayama A, Parker JA, Mihaljevic T, Laurence RG, Dor DM, Cohn LH, Bawendi MG, Frangioni JV. 2004.Near-infrared fluorescent type II quantum dots for sentinel lymph node mapping. Nat Biotechnol;22(1):93-7. Kim SW, Zimmer JP, Ohnishi S, Tracy JB, Frangioni JV, Bawendi MG. 2005.Engineering InAs(x)P(1-x)/InP/ZnSe III-V alloyed core/shell quantum dots for the near-infrared. J Am Chem Soc;127(30):10526-32. Kircheis R, Wightman L, Wagner E. 2001.Design and gene delivery activity of modified polyethylenimines. Advanced Drug Delivery Reviews;53(3):341-58. Klemm AR, Young D, Lloyd JB. 1998.Effects of polyethyleneimine on endocytosis and lysosome stability. Biochem Pharmacol;56(1):41-6. Kobayashi S, Hiroishi K, Tokunoh M, Saegusa T. 1987.Chelating Properties Of Linear And Branched Poly(Ethylenimines). Macromolecules;20(7):1496-500. 145 _______________________________________________________________________________ Konan YN, Gurny R, Allemann E. 2002.State of the art in the delivery of photosensitizers for photodynamic therapy. J Photochem Photobiol B;66(2):89-106. Konig K. 2000.Multiphoton microscopy in life sciences. Journal Of MicroscopyOxford;200(83-104. Kubitschko S, Spinke J, Bruckner T, Pohl S, Oranth N. 1997.Sensitivity enhancement of optical immunosensors with nanoparticles. Anal Biochem;253(1):112-22. Kuningas K, Rantanen T, Karhunen U, Lovgren T, Soukka T. 2005.Simultaneous use of time-resolved fluorescence and anti-stokes photoluminescence in a bioaffinity assay. Anal Chem;77(9):2826-34. Kuningas K, Rantanen T, Ukonaho T, Lovgren T, Soukka T. 2005.Homogeneous assay technology based on upconverting phosphors. Analytical Chemistry;77(22):7348-55. Lambert G, Bertrand JR, Fattal E, Subra F, Pinto-Alphandary H, Malvy C, Auclair C, Couvreur P. 2000.EWS fli-1 antisense nanocapsules inhibits ewing sarcomarelated tumor in mice. Biochem Biophys Res Commun;279(2):401-6. Lambert RC, Maulet Y, Dupont JL, Mykita S, Craig P, Volsen S, Feltz A. 1996.Polyethylenimine-mediated DNA transfection of peripheral and central neurons in primary culture: probing Ca2+ channel structure and function with antisense oligonucleotides. Mol Cell Neurosci;7(3):239-46. Larson DR, Zipfel WR, Williams RM, Clark SW, Bruchez MP, Wise FW, Webb WW. 2003.Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science;300(5624):1434-6. LeBien TW, McCormack RT. 1989.The common acute lymphoblastic leukemia antigen (CD10)--emancipation from a functional enigma. Blood;73(3):625-35. Leuschner C, Kumar CSSR. Nanoparticles for Cancer Drug delivery. In: Kumar C.S.S.R. HJ, Leuschner C., ed. Nanofabrication Towards Biomedical Applications. Verlag: Wiley-VCH, 2005:289 - 326. Lim SF, Riehn R, Ryu WS, Khanarian N, Tung CK, Tank D, Austin RH. 2006.In vivo and scanning electron microscopy imaging of up-converting nanophosphors in Caenorhabditis elegans. Nano Lett;6(2):169-74. Lvov Y, Ariga K, Ichinose I, Kunitake T. 1995.Assembly Of Multicomponent Protein Films By Means Of Electrostatic Layer-By-Layer Adsorption. Journal Of The American Chemical Society;117(22):6117-23. Mattes MJ, Major PP, Goldenberg DM, Dion AS, Hutter RV, Klein KM. 1990.Patterns of antigen distribution in human carcinomas. Cancer Res;50(3 Suppl):880s-4s. McNeil SE. 2005.Nanotechnology for the biologist. J Leukoc Biol. 146 _______________________________________________________________________________ Medintz IL, Uyeda HT, Goldman ER, Mattoussi H. 2005.Quantum dot bioconjugates for imaging, labelling and sensing. Nature Materials;4(6):435-46. Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, Sundaresan G, Wu AM, Gambhir SS, Weiss S. 2005.Quantum dots for live cells, in vivo imaging, and diagnostics. Science;307(5709):538-44. Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, Sundaresan G, Wu AM, Gambhir SS, Weiss S. 2005.Quantum dots for live cells, in vivo imaging, and diagnostics. Science;307(5709):538-44. Morgan NY, English S, Chen W, Chernomordik V, Russo A, Smith PD, Gandjbakhche A. 2005.Real time in vivo non-invasive optical imaging using near-infrared fluorescent quantum dots. Acad Radiol;12(3):313-23. Morin C, Barratt G, Fessi H, Devissaguet JP, Puisieux F. 1994.Improved intracellular delivery of a muramyl dipeptide analog by means of nanocapsules. Int J Immunopharmacol;16(5-6):451-6. Netti PA, Baxter LT, Boucher Y, Skalak R, Jain RK. 1995.Time-dependent behavior of interstitial fluid pressure in solid tumors: implications for drug delivery. Cancer Res;55(22):5451-8. Niedbala RS, Feindt H, Kardos K, Vail T, Burton J, Bielska B, Li S, Milunic D, Bourdelle P, Vallejo R. 2001.Detection of analytes by immunoassay using upconverting phosphor technology. Anal Biochem;293(1):22-30. Niedbala RS, Feindt H, Kardos K, Vail T, Burton J, Bielska B, Li S, Milunic D, Bourdelle P, Vallejo R. 2001.Detection of analytes by immunoassay using upconverting phosphor technology. Analytical Biochemistry;293(1):22-30. Oyewumi MO, Yokel RA, Jay M, Coakley T, Mumper RJ. 2004.Comparison of cell uptake, biodistribution and tumor retention of folate-coated and PEG-coated gadolinium nanoparticles in tumor-bearing mice. J Control Release;95(3):61326. Palmer RJ, Butenhoff JL, Stevens JB. 1987.Cytotoxicity of the rare earth metals cerium, lanthanum, and neodymium in vitro: comparisons with cadmium in a pulmonary macrophage primary culture system. Environ Res;43(1):142-56. Parungo CP, Ohnishi S, Kim SW, Kim S, Laurence RG, Soltesz EG, Chen FY, Colson YL, Cohn LH, Bawendi MG, Frangioni JV. 2005.Intraoperative identification of esophageal sentinel lymph nodes with near-infrared fluorescence imaging. J Thorac Cardiovasc Surg;129(4):844-50. Pires AM, Heer S, Gudel HU, Serra OA. 2006.Er, Yb doped yttrium based nanosized phosphors: particle size, "host lattice" and doping ion concentration effects on upconversion efficiency. J Fluoresc;16(3):461-8. Potineni A, Lynn DM, Langer R, Amiji MM. 2003.Poly(ethylene oxide)-modified poly(beta-amino ester) nanoparticles as a pH-sensitive biodegradable system for paclitaxel delivery. J Control Release;86(2-3):223-34. 147 _______________________________________________________________________________ Quintana A, Raczka E, Piehler L, Lee I, Myc A, Majoros I, Patri AK, Thomas T, Mule J, Baker JR, Jr. 2002.Design and function of a dendrimer-based therapeutic nanodevice targeted to tumor cells through the folate receptor. Pharm Res;19(9):1310-6. Ricci-Junior E, Marchetti JM. 2006.Preparation, characterization, photocytotoxicity assay of PLGA nanoparticles containing zinc (II) phthalocyanine for photodynamic therapy use. J Microencapsul;23(5):523-38. Ross JF, Chaudhuri PK, Ratnam M. 1994.Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications. Cancer;73(9):2432-43. Santra S, Liesenfeld B, Dutta D, Chatel D, Batich CD, Tan W, Moudgil BM, Mericle RA. 2005.Folate conjugated fluorescent silica nanoparticles for labeling neoplastic cells. J Nanosci Nanotechnol;5(6):899-904. Schwab G, Chavany C, Duroux I, Goubin G, Lebeau J, Helene C, Saison-Behmoaras T. 1994.Antisense oligonucleotides adsorbed to polyalkylcyanoacrylate nanoparticles specifically inhibit mutated Ha-ras-mediated cell proliferation and tumorigenicity in nude mice. Proc Natl Acad Sci U S A;91(22):10460-4. Shenoy DB, Amiji MM. 2005.Poly(ethylene oxide)-modified poly(epsiloncaprolactone) nanoparticles for targeted delivery of tamoxifen in breast cancer. Int J Pharm;293(1-2):261-70. Sinek J, Frieboes H, Zheng X, Cristini V. 2004.Two-dimensional chemotherapy simulations demonstrate fundamental transport and tumor response limitations involving nanoparticles. Biomed Microdevices;6(4):297-309. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. 1987.Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science;235(4785):177-82. Sledge GW, Jr., Miller KD. 2003.Exploiting the hallmarks of cancer: the future conquest of breast cancer. Eur J Cancer;39(12):1668-75. So MK, Xu C, Loening AM, Gambhir SS, Rao J. 2006.Self-illuminating quantum dot conjugates for in vivo imaging. Nat Biotechnol;24(3):339-43. Soltesz EG, Kim S, Kim SW, Laurence RG, De Grand AM, Parungo CP, Cohn LH, Bawendi MG, Frangioni JV. 2006.Sentinel lymph node mapping of the gastrointestinal tract by using invisible light. Ann Surg Oncol;13(3):386-96. Soltesz EG, Kim S, Laurence RG, DeGrand AM, Parungo CP, Dor DM, Cohn LH, Bawendi MG, Frangioni JV, Mihaljevic T. 2005.Intraoperative sentinel lymph node mapping of the lung using near-infrared fluorescent quantum dots. Ann Thorac Surg;79(1):269-77; discussion -77. Soukka T, Kuningas K, Rantanen T, Haaslahti V, Lovgren T. 2005.Photochemical characterization of up-converting inorganic lanthanide phosphors as potential labels. J Fluoresc;15(4):513-28. 148 _______________________________________________________________________________ Soukka T, Kuningas K, Rantanen T, Haaslahti V, Lovgren T. 2005.Photochemical characterization of up-converting inorganic lanthanide phosphors as potential labels. Journal Of Fluorescence;15(4):513-28. Taylor-Papadimitriou J, Burchell J, Miles DW, Dalziel M. 1999.MUC1 and cancer. Biochim Biophys Acta;1455(2-3):301-13. Teicher BA. 2000.Molecular targets and cancer therapeutics: discovery, development and clinical validation. Drug Resist Updat;3(2):67-73. Thorne RG, Nicholson C. 2006.In vivo diffusion analysis with quantum dots and dextrans predicts the width of brain extracellular space. Proc Natl Acad Sci U S A;103(14):5567-72. Toffoli G, Cernigoi C, Russo A, Gallo A, Bagnoli M, Boiocchi M. 1997.Overexpression of folate binding protein in ovarian cancers. Int J Cancer;74(2):193-8. van de Rijke F, Zijlmans H, Li S, Vail T, Raap AK, Niedbala RS, Tanke HJ. 2001.Up-converting phosphor reporters for nucleic acid microarrays. Nature Biotechnology;19(3):273-6. van De Rijke F, Zijlmans H, Li S, Vail T, Raap AK, Niedbala RS, Tanke HJ. 2001.Up-converting phosphor reporters for nucleic acid microarrays. Nat Biotechnol;19(3):273-6. Vervoordeldonk SF, Merle PA, van Leeuwen EF, von dem Borne AE, SlaperCortenbach IC. 1994.Preclinical studies with radiolabeled monoclonal antibodies for treatment of patients with B-cell malignancies. Cancer;73(3 Suppl):1006-11. Voura EB, Jaiswal JK, Mattoussi H, Simon SM. 2004.Tracking metastatic tumor cell extravasation with quantum dot nanocrystals and fluorescence emissionscanning microscopy. Nat Med;10(9):993-8. Wang F, Chatterjee DK, Li Z, Zhang Y, Fan X, Wang M. 2006.Synthesis of polyethylenimine/NaYF44 nanoparticles with upconversion fluorescence. Nanotechnology;17(23):5786. Wang F, Chatterjee DK, Li Z, Zhang Y, Fan XP, Wang MQ. 2006.Synthesis of polyethylenimine/NaYF44 nanoparticles with upconversion fluorescence. Nanotechnology;In Press( Wang F, Tan WB, Zhang Y, Fan XP, Wang MQ. 2006.Luminescent nanomaterials for biological labelling. Nanotechnology;17(R1-R13. Wang J, Chen Z, Corstjens PL, Mauk MG, Bau HH. 2006.A disposable microfluidic cassette for DNA amplification and detection. Lab Chip;6(1):46-53. Wang LY, Yan RX, Hao ZY, Wang L, Zeng JH, Bao H, Wang X, Peng Q, Li YD. 2005.Fluorescence resonant energy transfer biosensor based on upconversion- 149 _______________________________________________________________________________ luminescent nanoparticles. Angewandte Chemie-International Edition;44(37):6054-7. Wang X, Zhuang J, Peng Q, Li Y. 2006.Hydrothermal synthesis of rare-earth fluoride nanocrystals. Inorg Chem;45(17):6661-5. Weitman SD, Frazier KM, Kamen BA. 1994.The folate receptor in central nervous system malignancies of childhood. J Neurooncol;21(2):107-12. Weitman SD, Lark RH, Coney LR, Fort DW, Frasca V, Zurawski VR, Jr., Kamen BA. 1992.Distribution of the folate receptor GP38 in normal and malignant cell lines and tissues. Cancer Res;52(12):3396-401. Weitman SD, Weinberg AG, Coney LR, Zurawski VR, Jennings DS, Kamen BA. 1992.Cellular localization of the folate receptor: potential role in drug toxicity and folate homeostasis. Cancer Res;52(23):6708-11. Wieder ME, Hone DC, Cook MJ, Handsley MM, Gavrilovic J, Russell DA. 2006.Intracellular photodynamic therapy with photosensitizer-nanoparticle conjugates: cancer therapy using a 'Trojan horse'. Photochem Photobiol Sci;5(8):727-34. Wu X, Liu H, Liu J, Haley KN, Treadway JA, Larson JP, Ge N, Peale F, Bruchez MP. 2003.Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots. Nat Biotechnol;21(1):41-6. Yamamoto T, Ikawa S, Akiyama T, Semba K, Nomura N, Miyajima N, Saito T, Toyoshima K. 1986.Similarity of protein encoded by the human c-erb-B-2 gene to epidermal growth factor receptor. Nature;319(6050):230-4. Yi GS, Lu HC, Zhao SY, Yue G, Yang WJ, Chen DP, Guo LH. 2004.Synthesis, characterization, and biological application of size-controlled nanocrystalline NaYF44: Yb,Er infrared-to-visible up-conversion phosphors. Nano Letters;4(11):2191-6. Yi GS, Sun BQ, Yang FZ, Chen DP, Zhou YX, Cheng J. 2002.Synthesis and characterization of high-efficiency nanocrystal up-conversion phosphors: Ytterbium and erbium codoped lanthanum molybdate. Chemistry Of Materials;14(7):2910-4. Zeng JH, Su J, Li ZH, Yan RX, Li YD. 2005.Synthesis and upconversion luminescence of hexagonal-phase NaYF44: Yb, Er3+, phosphors of controlled size and morphology. Advanced Materials;17(17):2119-23. Zhang P, Rogelj S, Nguyen K, Wheeler D. 2006.Design of a highly sensitive and specific nucleotide sensor based on photon upconverting particles. J Am Chem Soc;128(38):12410-1. Zhang P, Steelant W, Kumar M, Scholfield M. 2007.Versatile photosensitizers for photodynamic therapy at infrared excitation. J Am Chem Soc;129(15):4526-7. 150 _______________________________________________________________________________ Zhang Y, Chatterjee DK. Liposomes, dendrimers and other polymeric nanoparticles for targeted delivery of anticancer agents - A comparative study. In: Kumar CSSR, ed. Nanomaterials for Cancer Therapy. Weinheim: Wiley-VCH Verlag GmbH & Co, KGaA, 2006:338 - 70. Zijlmans H, Bonnet J, Burton J, Kardos K, Vail T, Niedbala RS, Tanke HJ. 1999.Detection of cell and tissue surface antigens using up-converting phosphors: A new reporter technology. Analytical Biochemistry;267(1):30-6. 151 [...]... nanoparticles in tumors is followed by sections which discuss fluorescent nanoparticles used in diagnosis and drug-loaded nanoparticles in therapy 3 _ 1.2.1 Molecular targeting using nanoparticles Definition: Targeting can be loosely defined in this context as any means that increases the specificity of localization of nanoparticles to diseased cells Targeting does not intrinsically... explored biomedical applications of nanoparticles Ferrari (Ferrari, 2005) has dealt with the whole field of cancer nanotechnology, including the in vitro diagnostics as well as in vivo targeting, while Jain has focused on drug delivery in cancer (Jain, 2005) Others have discussed nanotechnology for the biologist (McNeil, 2005) and its uses to the whole field of molecular recognition, mainly for enhanced in. .. either increasing concentration of particles, or by using larger particles or by using higher power of incident radiation The process of upconversion does not necessarily require coherent light, but a focused laser beam is often used in order to obtain high emission efficiency The remarkable photostability of the particles allows the use of laser for relatively long periods without significant bleaching... concentration of the toxic drug while reducing circulating levels of free drug, thus lowering systemic toxicity (Leuschner et al., 2005) Figure 1-1 Different roles of nanoparticles for biomedical applications The following sections deal with each of the three functions separately with specific reference to application in cancer diagnosis and management Description of methods of targeting and concentrating nanoparticles. .. Figure 4-4 Comparing emission from subcutaneous injections of QDs and UCN .111 xiv _ Figure 4-5 Injection into other tissues in Wistar rats: A) muscle and B) heart showed detectable fluorescence 113 Figure 4-6 In vivo live cell imaging using NaYF4 nanoparticles The ‘floating balls’ appearance of the cells in the bottom panel has a distinctive 3-dimensional... Figure 1-2 Tumor targeting with nanoparticles by passive and active targeting 5 Figure 1-3 Principle of photodynamic therapy 20 Figure 1-4 Upconversion involves energy transfer between two excited ion species, resulting in the acceptor ions reaching a higher energy state and subsequently emitting higher energy radiation In contrast, single photon fluorescence has emission of lower energy ... above, the following discussions relate specifically to tumors Passive targeting involves modifications of nanoparticles which increase circulation time without addition of any component/involvement of any method which is specific to the tumor Increased circulation time helps in accumulation of the particles in the tumor by an enhanced permeation and retention, or EPR, effect Long circulating nanoparticles. .. form of special polymer coatings which use steric stabilization The resultant nanoparticles are named ‘stealth’ 5 _ nanoparticles For example, attachment of poly(ethylene glycol) (PEG) ‘hides’ nanoparticles from the MPS enabling longer circulation times Longer time in circulation increases the probability of the nanoparticles being trapped in tumors by EPS In fact,... PEG-coated poly(cyanoacrylate)(pCA) nanoparticles - made by a copolymer inculcating both – has such a long circulating time that they penetrated the brain more than any other modifications, including coating by polysorbate This uptake was increased in pathological situations with presumably higher blood-brain barrier permeability Another example is the incorporation of cisplatin in liposomal formulations (Chawla... with target molecules causes unpredictable changes in the molecular structure of the organic fluorophore resulting in variable reduction in fluorescence conversion efficiency 9 _ Quantum dots (QDs) are nanoparticles of one semiconductor encased in a second semiconductor These can be of considerable use as inorganic fluorophores, because they offer significant advantages . subcutaneous injection of nanoparticles 111 4.3.3 Imaging of injected nanoparticles in other tissues 112 4.3.4 In vivo cell imaging 113 4.4. Conclusion 116 CHAPTER 5 UPCONVERSION NANOPARTICLES IN. Proceedings of The 12th International Conference on Biomedical Engineering (2005). Singapore: IFMBE. (The 12th International Conference on Biomedical Engineering (ICBME 2005), 7 - 10 Dec 2005, Singapore). upconversion nanoparticles with QDs for in vivo imaging 105 4.2.4 Imaging of upconversion nanoparticles in other rat tissues 106 4.2.5 In vivo microscopy using upconversion nanoparticles

Ngày đăng: 14/09/2015, 14:12

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan