• Keine Ergebnisse gefunden

Abs Adsorbed Abs

4.3. Adsorption of gold nanoparticles to hydroxyapatite

0 0.1 0.2 0.3 0.4 0.5 0.6 0.7

400 500 600 700 800 900 1000

Wave length (nm)

Absorbance

Figure 7: Effect of bovine serum (10 % v/v) on the UV-Vis absorption spectra of mPEG-AlkSH, BP-PEG-AlkSH and citrate coated gold nanoparticles (from top to bottom)

4.3. Adsorption of gold nanoparticles to hydroxyapatite

The adsorption of bisphosphonate modified GNPs (GNPs-BP) to hydroxyapatite was studied to evaluate the affinity of surface-modified gold nanoparticles to bone mineral. For this study endobone was used as a porous hydroxyapatite ceramic produced by sintering of bovine bone and it consequently can be used as model of bone, because it is the major constituent of the natural bone besides the protein [42]. The hydroxyapatite binding of GNPs modified with different amounts of bisphosphonate in the polymer layer was compared with the binding of GNPs coated with 100% of mPEG-AlkSH (0% bisphosphonate). The results showed that increasing the amount of binding ligand for the hydroxyapatite surface (bisphosphonate) led to increasing amounts of GNPs adsorbed to hydroxyapatite as depicted in Figure 8.

0 20 40 60 80 100 120

0% 25% 50% 75% 100%

Bisphosphonate conc. (%)

% Binding to HA

1 day 2 days 3 days 4 days

Figure 8: Hydroxyapatite binding of GNPs modified with different amounts of bisphosphonate (e.g, 75% means that GNPs are coated with 75% BP-PEG-AlkSH and 25% mPEG-AlkSH)

GNPs modified with 0% Bp (100% mPEG-AlkSH) showed no affinity to hydroxyapatite, while about 25% of the initially added GNPs modified with 25% BP (75% mPEG-AlkSH) adsorbed to hydroxyapatite within 24 hours. This adsorbed amount increased to about 40% after 48 hours with no further increase in the adsorbed amount by shaking for longer time (up to 4 days). The bound amounts of particles modified with 50% bisphosphonate were about 45% after one day and reached 64% after two days. The adsorbed amounts of GNPs modified with 75% and 100% bisphosphonate were 58% and 37% after one day, and 76% and 81% after two days and reached 86% and 100% after four days respectively. The steady increase in the adsorbed amounts with increasing concentration of bisphosphonate in the particle surface can be explained by the higher hydroxyapatite affinity of GNPs containing more bisphosphonate in their surface. The relatively long time (about two days) to establish the equilibrium between the adsorbed and free particles can be attributed to the necessary diffusion of the particles within the large volume of

155

the dispersion medium and the necessary penetration into the macro and micro pores of endobone granules on the bottom of the flasks to reach the adsorption sites. Similar results are obtained by many other authors who studied the binding of nanoparticles to hydroxyapatite [55,60].

Since it is well known that bisphosphonate strongly binds to divalent ions, such as calcium [61], the binding assay of bisphosphonate-modified GNPs was repeated in presence of calcium chloride to explore effect of calcium ions on the affinity of the nanoparticles. The used calcium concentration was 2.5 mM, which is the physiological concentration present in blood serum. The binding affinity of 100% bisphosphonate-modified GNPs to hydroxyapatite in the presence and absence of calcium chloride is depicted in Figure 9. It could be observed that there is no difference in the binding of nanoparticles either in presence or absence of calcium. This indicates that the binding affinity of particles to hydroxyapatite is not affected by the presence of calcium ions in the colloidal solution.

0 20 40 60 80 100 120

24 48

Time (hours)

% Binding to HA

GNP-BP GNP-BP + CaCl2

Figure 9: Effect of calcium chloride on the hydroxyapatite binding of bisphosphonate-modified GNPs

Furthermore, the in-vitro binding of bisphosphonate-modified GNPs was carried out in presence of BSA (200 µg/ml) and serum (10% v/v) to determine, if the interaction of bisphosphonate with these proteins compromises the binding to hydroxyapatite. The results of these experiments are presented in Figure 10, which shows that the binding affinity of nanoparticles to hydroxyapatite is not affected by the presence of BSA or serum. The obtained data indicate that the bisphosphonate-modified GNPs are stable and the observed interaction of particles and proteins, especially BSA (Figure 5), does not influence their bone mineral affinity.

0 20 40 60 80 100 120

GNPs-BP GNPs-BP + BSA GNPs-BP + Serum

% Binding to HA

2 days 4 days

Figure 10: Effect of BSA (200 µg/ml) and bovine serum (10% v/v) on the binding of bisphosphonate-modified GNPs to hydroxyapatite

157

5. Conclusions

In conclusion, it could be shown that the synthesized polymers, methoxy poly(ethylene glycol)-undecyl mercaptane and the bisphosphonate derivative, are suitable to functionalize gold nanoparticles prepared by citrate reduction. The obtained particles were characterized with respect to size, surface charges and dispersion stability. The polymer modified GNPs showed an enhanced stability against aggregation in different solutions such as high ionic strength NaCl solutions, as well as solutions containing BSA and serum.

Furthermore, the modification of the particles with bisphosphonate modified polymers allowed the preparation of particles with high affinity and strong binding to hydroxyapatite. The binding affinity to bone mineral could be adjusted by increasing the amount of bisphosphonate (targeting ligand) on the particle surfaces. Furthermore, it was demonstrated that the binding is not affected by present calcium ions as well as in presence of proteins.

Based on the conducted in vitro studies, bisphosphonate-functionalized GNPs seem to be a promising model for the investigation of the in vivo bone targeting of nanoparticles. An adjustment of the binding affinity can be achieved by changing the amounts of immobilized bisphosphonate modified polymers. An exchange for other suitable ligands allows using the nanoparticle system also for other targets and the possible exploration for other applications than bone.

6. References

1. Anil K. Patri, Jolanta F. Kukowska-Latallo, and James R. Baker, "Targeted drug delivery with dendrimers: Comparison of the release kinetics of covalently conjugated drug and non-covalent drug inclusion complex," Advanced Drug Delivery Reviews 57, 2203-2214 (2005).

2. Jolanta F. Kukowska-Latallo, Kimberly A. Candido, Zhengyi Cao, Shraddha S. Nigavekar, Istvan J. Majoros, Thommey P. Thomas, Lajos P. Balogh, Mohamed K. Khan, and James R.

Baker, Jr., "Nanoparticle targeting of anticancer drug improves therapeutic response in animal model of human epithelial cancer," Cancer Res 65, 5317-5324 (2005).

3. Megan E. Pearce, Jessica B. Melanko, and Aliasger K. Salem, "Multifunctional Nanorods for Biomedical Applications," Pharmaceutical Research 24, 2335-2352 (2007).

4. Lilian E. van Vlerken and Mansoor M. Amiji, "Multi-functional polymeric nanoparticles for tumour-targeted drug delivery," Expert Opinion on Drug Delivery 3, 205-216 (2006).

5. Eric Forssen and Michael Willis, "Ligand-targeted liposomes," Advanced Drug Delivery Reviews 29, 249-271 (1998).

6. Jessica B. Melanko, Megan E. Pearce, and Aliasger K. Salem, "Nanotubes, nanorods, nanofibers, and fullerenes for nanoscale drug delivery," Biotechnology: Pharmaceutical Aspects 10, 105-127 (2009).

7. Stuart C. McBain, Humphrey H. P. Yiu, and Jon Dobson, "Magnetic nanoparticles for gene and drug delivery," International Journal of Nanomedicine 3, 169-180 (2008).

8. Stephen Hanessian, Justyna A. Grzyb, Feride Cengelli, and Lucienne Juillerat-Jeanneret,

"Synthesis of chemically functionalized superparamagnetic nanoparticles as delivery vectors for chemotherapeutic drugs," Bioorganic & Medicinal Chemistry 16, 2921-2931 (2008).

9. O. V. Salata, "Applications of nanoparticles in biology and medicine," Journal of Nanobiotechnology 2, No (2004).

10. Ximei Qian, Xiang Hong Peng, Dominic O. Ansari, Qiqin Yin-Goen, Georgia Z. Chen, Dong M. Shin, Lily Yang, Andrew N. Young, May D. Wang, and Shuming Nie, "In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags,"

Nature Biotechnology 26, 83-90 (2008).

11. Yanli Liu and Stefan Franzen, "Factors Determining the Efficacy of Nuclear Delivery of Antisense Oligonucleotides by Gold Nanoparticles," Bioconjugate Chem. 19, 1009-1016 (2008).

12. Giulio F. Paciotti, Lonnie Myer, David Weinreich, Dan Goia, Nicolae Pavel, Richard E.

McLaughlin, and Lawrence Tamarkin, "Colloidal gold: a novel nanoparticle vector for tumor directed drug delivery," Drug delivery 11, 169-183 (2004).

13. Priyabrata Mukherjee, Resham Bhattacharya, Nancy Bone, Yean K. Lee, Chitta Ranjan Patra, Shanfeng Wang, Lichun Lu, Charla Secreto, Pataki C. Banerjee, Michael J. Yaszemski, Neil

159

E. Kay, and Debabrata Mukhopadhyay, "Potential therapeutic application of gold nanoparticles in B-chronic lymphocytic leukemia (BCLL): enhancing apoptosis," Journal of Nanobiotechnology 5, No (2007).

14. Rachana K. Visaria, Robert J. Griffin, Brent W. Williams, Emad S. Ebbini, Giulio F. Paciotti, Chang W. Song, and John C. Bischof, "Enhancement of tumor thermal therapy using gold nanoparticle-assisted tumor necrosis factor-a delivery," Molecular Cancer Therapeutics 5, 1014-1020 (2006).

15. Rachana Visaria, John C. Bischof, Melissa Loren, Brent Williams, Emad Ebbini, Giulio Paciotti, and Robert Griffin, "Nanotherapeutics for enhancing thermal therapy of cancer," Int J Hyperthermia 23, 501-511 (2007).

16. Yu Hung Chen, Chiau Yuang Tsai, Pon Yu Huang, Meng Ya Chang, Pai Chiao Cheng, Chen Hsi Chou, Dong Hwang Chen, Chrong Reen Wang, Ai Li Shiau, and Chao Liang Wu,

"Methotrexate Conjugated to Gold Nanoparticles Inhibits Tumor Growth in a Syngeneic Lung Tumor Model," Molecular Pharmaceutics 4, 713-722 (2007).

17. L. B. Nie, Y. Yang, S. Li, and N. Y. He, "Enhanced DNA detection based on the amplification of gold nanoparticles using quartz crystal microbalance," Nanotechnology 18, 305501-1-305501/5 (2007).

18. Grit Festag, Andrea Steinbrueck, Andreas Wolff, Andrea Csaki, Robert Moeller, and Wolfgang Fritzsche, "Optimization of gold nanoparticles-based DNA detection for microarrays," Journal of Fluorescence 15, 161-170 (2005).

19. Joong H. Kim, R. August Estabrook, Gary Braun, Briana R. Lee, and Norbert O. Reich,

"Specific and sensitive detection of nucleic acids and RNases using gold nanoparticle-RNA-fluorescent dye conjugates," Chemical Communications (Cambridge, United Kingdom) 4342-4344 (2007).

20. C. Shad Thaxton, Dimitra G. Georganopoulou, and Chad A. Mirkin, "Gold nanoparticle probes for the detection of nucleic acid targets," Clinica Chimica Acta 363, 120-126 (2006).

21. Charng Sheng Tsai, Ting Bin Yu, and Chao Tsen Chen, "Gold nanoparticle-based competitive colorimetric assay for detection of protein-protein interactions," Chemical Communications (Cambridge, United Kingdom) 4273-4275 (2005).

22. Kadir Aslan, Jian Zhang, Joseph R. Lakowicz, and Chris D. Geddes, "Saccharide sensing using gold and silver nanoparticles - A review," Journal of Fluorescence 14, 391-400 (2004).

23. J. Ma, Huifen Wong, L. B. Kong, and K. W. Peng, "Biomimetic processing of nanocrystallite bioactive apatite coating on titanium," Nanotechnology 14, 619-623 (2003).

24. Agustin De La Isla, Witold Brostow, Bernard Bujard, Miriam Estevez, J. Rogelio Rodriguez, Susana Vargas, and Victor M. Castano, "Nanohybrid scratch resistant coatings for teeth and bone viscoelasticity manifested in tribology," Materials Research Innovations 7, 110-114 (2003).

25. Chenjie Xu, Glenn A. Tung, and Shouheng Sun, "Size and Concentration Effect of Gold Nanoparticles on X-ray Attenuation As Measured on Computed Tomography," Chemistry of Materials ACS (2008).

26. J. F. Hainfeld, D. N. Slatkin, T. M. Focella, and H. M. Smilowitz, "Gold nanoparticles: a new X-ray contrast agent," British Journal of Radiology 79, 248-253 (2006).

27. Jingyuan Li, Xuemei Wang, Chunxia Wang, Baoan Chen, Yongyuan Dai, Renyun Zhang, Min Song, Gang Lv, and Degang Fu, "The enhancement effect of gold nanoparticles in drug delivery and as biomarkers of drug-resistant cancer cells," ChemMedChem 2, 374-378 (2007).

28. Catherine M. Goodman, Catherine D. McCusker, Tuna Yilmaz, and Vincent M. Rotello,

"Toxicity of Gold Nanoparticles Functionalized with Cationic and Anionic Side Chains,"

Bioconjugate Chem. 15, 897-900 (2004).

29. Jun Shan and Heikki Tenhu, "Recent advances in polymer protected gold nanoparticles:

synthesis, properties and applications," Chemical Communications (Cambridge, United Kingdom) 4580-4598 (2007).

30. Ralph Weissleder, Kimberly Kelly, Eric Yi Sun, Timur Shtatland, and Lee Josephson, "Cell-specific targeting of nanoparticles by multivalent attachment of small molecules," Nature Biotechnology 23, 1418-1423 (2005).

31. Xiangyang Shi, Suhe Wang, Sasha Meshinchi, Mary E. Van Antwerp, Xiangdong Bi, Inhan Lee, and James R. Baker, Jr., "Dendrimer-entrapped gold nanoparticles as a platform for cancer-cell targeting and imaging," Small 3, 1245-1252 (2007).

32. Hidenori Otsuka, Yoshitsugu Akiyama, Yukio Nagasaki, and Kazunori Kataoka,

"Quantitative and reversible lectin-induced association of gold nanoparticles modified with a-lactosyl-w-mercapto-poly(ethylene glycol)," Journal of the American Chemical Society 123, 8226-8230 (2001).

33. A. H. Lu, E. L. Salabas, and F. Schueth, "Magnetic nanoparticles: synthesis, protection, functionalization, and application," Angewandte Chemie, International Edition 46, 1222-1244 (2007).

34. G. Frens, "Controlled nucleation for the regulation of the particle size in monodisperse gold suspensions," Nature (London), Physical Science 241, 20-22 (1973).

35. Katherine C. Grabar, R. Griffith Freeman, Michael B. Hommer, and Michael J. Natan,

"Preparation and Characterization of Au Colloid Monolayers," Analytical Chemistry 67, 735-743 (1995).

36. Yanli Liu, Mathew K. Shipton, Joseph Ryan, Eric D. Kaufman, Stefan Franzen, and Daniel L.

Feldheim, "Synthesis, Stability, and Cellular Internalization of Gold Nanoparticles Containing Mixed Peptide-Poly(ethylene glycol) Monolayers," Analytical Chemistry 79, 2221-2229 (2007).

161

37. M. Rahimnejad, M. Jahanshahi, and G. D. Najafpour, "Production of biological nanoparticles from bovine serum albumin for drug delivery," African Journal of Biotechnology 5, 1918-1923 (2006).

38. Andy Scheffer, Carsten Engelhard, Michael Sperling, and Wolfgang Buscher, "ICP-MS as a new tool for the determination of gold nanoparticles in bioanalytical applications," Analytical and Bioanalytical Chemistry 390, 249-252 (2008).

39. Claire Mangeney, Fabien Ferrage, Isabelle Aujard, Valerie Marchi-Artzner, Ludovic Jullien, Olivier Ouari, El Djouhar Rekaie, Andre Laschewsky, Inger Vikholm, and Janusz W.

Sadowski, "Synthesis and Properties of Water-Soluble Gold Colloids Covalently Derivatized with Neutral Polymer Monolayers," Journal of the American Chemical Society 124, 5811-5821 (2002).

40. Seiji Takae, Yoshitsugu Akiyama, Hidenori Otsuka, Teisaku Nakamura, Yukio Nagasaki, and Kazunori Kataoka, "Ligand density effect on biorecognition by PEGylated gold nanoparticles: regulated interaction of RCA120 lectin with lactose installed to the distal end of tethered PEG strands on gold surface," Biomacromolecules 6, 818-824 (2005).

41. N. Bouropoulos and J. Moradian-Oldak, "Analysis of hydroxyapatite surface coverage by amelogenin nanospheres following the langmuir model for protein adsorption," Calcified Tissue International 72, 599-603 (2003).

42. S. Joschek, B. Nies, R. Krotz, and A. Gopferich, "Chemical and physicochemical characterization of porous hydroxyapatite ceramics made of natural bone," Biomaterials 21, 1645-1658 (2000).

43. Wolfgang Haiss, Nguyen T. K. Thanh, Jenny Aveyard, and David G. Fernig, "Determination of Size and Concentration of Gold Nanoparticles from UV-Vis Spectra," Analytical Chemistry (Washington, DC, United States) 79, 4215-4221 (2007).

44. Zul Merican, Tara L. Schiller, Craig J. Hawker, Peter M. Fredericks, and Idriss Blakey, "Self-assembly and encoding of polymer-stabilized gold nanoparticles with surface-enhanced Raman reporter molecules," Langmuir 23, 10539-10545 (2007).

45. Dirk Eck, Christiane A. Helm, Norman J. Wagner, and Abraham Vaynberg, "Plasmon Resonance Measurements of the Adsorption and Adsorption Kinetics of a Biopolymer onto Gold Nanocolloids. [Erratum to document cited in CA134:213208]," Langmuir 23, 9522 (2007).

46. Vivechana Dixit, Jeroen Van den Bossche, Debra M. Sherman, David H. Thompson, and Ronald P. Andres, "Synthesis and Grafting of Thioctic Acid-PEG-Folate Conjugates onto Au Nanoparticles for Selective Targeting of Folate Receptor-Positive Tumor Cells,"

Bioconjugate Chem. 17, 603-609 (2006).

47. S. L. Cumberland and G. F. Strouse, "Analysis of the Nature of Oxyanion Adsorption on Gold Nanomaterial Surfaces," Langmuir 18, 269-276 (2002).

48. Xiong Liu, Mark Atwater, Jinhai Wang, and Qun Huo, "Extinction coefficient of gold nanoparticles with different sizes and different capping ligands," Colloids and Surfaces, B:

Biointerfaces 58, 3-7 (2007).

49. Marie Gaumet, Angelica Vargas, Robert Gurny, and Florence Delie, "Nanoparticles for drug delivery: The need for precision in reporting particle size parameters," European Journal of Pharmaceutics and Biopharmaceutics 69, 1-9 (2008).

50. Boru Zhu, Thomas Eurell, Rico Gunawan, and Deborah Leckband, "Chain-length dependence of the protein and cell resistance of oligo(ethylene glycol)-terminated self-assembled monolayers on gold," Journal of Biomedical Materials Research 56, 406-416 (2001).

51. Devika R. Bhumkar, Hrushikesh M. Joshi, Murali Sastry, and Varsha B. Pokharkar,

"Chitosan Reduced Gold Nanoparticles as Novel Carriers for Transmucosal Delivery of Insulin," Pharmaceutical Research 24, 1415-1426 (2007).

52. Jingfang Zhou, David A. Beattie, John Ralston, and Rossen Sedev, "Colloid Stability of Thymine-Functionalized Gold Nanoparticles," Langmuir 23, 12096-12103 (2007).

53. Ruxandra Gref, Gregory Miralles, and Edith Dellacherie, "Polyoxyethylene-coated nanospheres: effect of coating on zeta potential and phagocytosis," Polymer International 48, 251-256 (1999).

54. Muriel K. Corbierre, Neil S. Cameron, and R. Bruce Lennox, "Polymer-stabilized gold nanoparticles with high grafting densities," Langmuir 20, 2867-2873 (2004).

55. V. Hengst, C. Oussoren, T. Kissel, and G. Storm, "Bone targeting potential of bisphosphonate-targeted liposomes," International Journal of Pharmaceutics 331, 224-227 (2007).

56. Saeeda Jaffar, Ki Tae Nam, Ali Khademhosseini, Jia Xing, Robert S. Langer, and Angela M.

Belcher, "Layer-by-layer surface modification and patterned electrostatic deposition of quantum dots," Nano Letters 4, 1421-1425 (2004).

57. Gang Wang and Wenfang Sun, "Optical Limiting of Gold Nanoparticle Aggregates Induced by Electrolytes," Journal of Physical Chemistry B 110, 20901-20905 (2006).

58. Santosh Aryal, K. C. R. Bahadur, Narayan Bhattarai, Chul Ki Kim, and Hak Yong Kim,

"Study of electrolyte induced aggregation of gold nanoparticles capped by amino acids,"

Journal of Colloid and Interface Science 299, 191-197 (2006).

59. R. L. C. Wang, H. J. Kreuzer, and M. Grunze, "Molecular Conformation and Solvation of Oligo(ethylene glycol)-Terminated Self-Assembled Monolayers," Journal of Physical Chemistry B 101, 9767-9773 (1997).

60. Sung Wook Choi and Jung Hyun Kim, "Design of surface-modified poly(d,l-lactide-co-glycolide) nanoparticles for targeted drug delivery to bone," Journal of Controlled Release 122, 24-30 (2007).

163

61. Anke J. Roelofs, Keith Thompson, Sharon Gordon, and Michael J. Rogers, "Molecular Mechanisms of Action of Bisphosphonates: current Status," Clinical Cancer Research 12, 6222s-6230s (2006).

Chapter 6

Targeting of Bisphosphonate-Functionlized