全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Nanomaterials  2013 

Investigation of Sub-100 nm Gold Nanoparticles for Laser-Induced Thermotherapy of Cancer

DOI: 10.3390/nano3010086

Keywords: gold nanorods, gold core-corona nanoparticles, hollow gold nanoshells, light scattering microscopy, prostate cancer, gene therapy

Full-Text   Cite this paper   Add to My Lib

Abstract:

Specialized gold nanostructures are of interest for the development of alternative treatment methods in medicine. Photothermal therapy combined with gene therapy that supports hyperthermia is proposed as a novel multimodal treatment method for prostate cancer. In this work, photothermal therapy using small (<100 nm) gold nanoparticles and near-infrared (NIR) laser irradiation combined with gene therapy targeting heat shock protein (HSP) 27 was investigated. A series of nanoparticles: nanoshells, nanorods, core-corona nanoparticles and hollow nanoshells, were synthesized and examined to compare their properties and suitability as photothermal agents. In vitro cellular uptake studies of the nanoparticles into prostate cancer cell lines were performed using light scattering microscopy to provide three-dimensional (3D) imaging. Small gold nanoshells (40 nm) displayed the greatest cellular uptake of the nanoparticles studied and were used in photothermal studies. Photothermal treatment of the cancer cell lines with laser irradiation at 800 nm at 4 W on a spot size of 4 mm (FWHM) for 6 or 10 min resulted in an increase in temperature of ~12 °C and decrease in cell viability of up to 70%. However, in vitro studies combining photothermal therapy with gene therapy targeting HSP27 did not result in additional sensitization of the prostate cancer cells to hyperthermia.

References

[1]  Scher, H.I.; Leiber, S.A.; Fuks, Z.; Cordon-Cardo, C.; Scardino, P.T. Cancer of The Prostate. In Cancer: Principles and Practice of Oncology, 7th; DeVita, V.T., Hellman, S., Rosenberg, S.A., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2005; pp. 1192–1259.
[2]  Zacharakis, E.; Ahmed, H.U.; Ishaq, A.; Scott, R.; Illing, R.; Freeman, A.; Allen, C.; Emberton, M. The feasibility and safety of high-intensity focused ultrasound as salvage therapy for recurrent prostate cancer following external beam radiotherapy. BJU Int. 2008, 102, 786–792, doi:10.1111/j.1464-410X.2008.07775.x.
[3]  Marberger, M.; Carroll, P.R.; Zelefsky, M.J.; Coleman, J.A.; Hricak, H.; Scardino, P.T.; Abenhaim, L.L. New treatments for localized prostate cancer. Urology 2008, 72, 36–43.
[4]  Lu, W.; Singh, A.K.; Khan, S.A.; Senapati, D.; Yu, H.; Ray, P.C. Gold nano-popcorn-based targeted diagnosis, nanotherapy treatment, and in situ monitoring of photothermal therapy response of prostate cancer cells using surface-enhanced raman spectroscopy. J. Am. Chem. Soc. 2010, 132, 18103–18114.
[5]  Canadian Cancer Society. Prostate Cancer Statistics. Available online: http://www.cancer.ca/British%20Columbia-Yukon/About%20cancer/Cancer%20statistics/Stats%20at%20a%20glance/Prostate%20cancer.aspx?sc_lang=en&r=1 (accessed on 12 October 2010).
[6]  Albertsen, P.C. Treatment of localized prostate cancer: When is active surveillance appropriate? Nat. Rev. Clin. Oncol. 2010, 7, 394–400, doi:10.1038/nrclinonc.2010.63.
[7]  Kirui, D.K.; Rey, D.A.; Batt, C.A. Gold hybrid nanoparticles for targeted phototherapy and cancer imaging. Nanotechnology 2010, 21, 105105–105115, doi:10.1088/0957-4484/21/10/105105.
[8]  Dickerson, E.B.; Dreaden, E.C.; Huang, X.H.; El-Sayed, I.H.; Chu, H.H.; Pushpanketh, S.; McDonald, J.F.; El-Sayed, M.A. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett. 2008, 269, 57–66, doi:10.1016/j.canlet.2008.04.026.
[9]  Hirsch, L.R.; Stafford, R.J.; Bankson, J.A.; Sershen, S.R.; Rivera, B.; Price, R.E.; Hazle, J.D.; Halas, N.J.; West, J.L. Nanoshell-mediated near-infrared thermal therapy of tumours under magnetic resonance guidance. Proc. Natl. Acad. Sci. USA 2003, 100, 13549–13554.
[10]  Khlebtsov, B.; Zharov, V.; Melnikov, A.; Tuchin, V.; Khlebtsov, N. Optical amplification of photothermal therapy with gold nanoparticles and nanoclusters. Nanotechnology 2006, 17, 5167–5179.
[11]  Weissleder, R. A clearer vision for in vivo imaging. Nat. Biotechnol. 2001, 19, 316–317.
[12]  Richardson, H.H.; Carlson, M.T.; Tandler, P.J.; Hernandez, P.; Govorov, A.O. Experimental and theoretical studies of light-to-heat conversion and collective heating effects in metal nanoparticle solutions. Nano Lett. 2009, 9, 1139–1146, doi:10.1021/nl8036905.
[13]  Jain, P.K.; Lee, K.S.; El-Sayed, I.H.; El-Sayed, M.A. Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: Applications in biological imaging and biomedicine. J. Phys. Chem. B 2006, 110, 7238–7248.
[14]  Kelly, K.L.; Coronado, E.; Zhao, L.L.; Schatz, G.C. The optical properties of metal nanoparticles: The influence of size, shape, and dielectric environment. J. Phys. Chem. B 2003, 107, 668–677.
[15]  Huang, X.H.; Jain, P.K.; El-Sayed, I.H.; El-Sayed, M.A. Plasmonic photothermal therapy (PPTT) using gold nanoparticles. Lasers Med. Sci. 2008, 23, 217–228, doi:10.1007/s10103-007-0470-x.
[16]  Niidome, T.; Yamagata, M.; Okamoto, Y.; Akiyama, Y.; Takahashi, H.; Kawano, T.; Katayama, Y.; Niidome, Y. PEG-modified gold nanorods with a stealth character for in vivo applications. J. Control. Release 2006, 114, 343–347, doi:10.1016/j.jconrel.2006.06.017.
[17]  Chen, J.Y.; Wang, D.L.; Xi, J.F.; Au, L.; Siekkinen, A.; Warsen, A.; Li, Z.Y.; Zhang, H.; Xia, Y.N.; Li, X.D. Immuno gold nanocages with tailored optical properties for targeted photothermal destruction of cancer cells. Nano Lett. 2007, 7, 1318–1322, doi:10.1021/nl070345g.
[18]  Erickson, T.A.; Tunnel, J.W. Gold Nanoshells in Biomedical Applications. In Mixed Metal Nanomaterials; Kumar, C.S.S.R., Ed.; Wiley-VCH: Weinheim, Germany, 2009; pp. 1–44.
[19]  Nanospectra Biosciences Inc. Tumour Ablation Using AuroLase? Therapy. Available online: http://www.nanospectra.com (accessed on 14 August 2010).
[20]  Prevo, B.G.; Esakoff, S.A.; Mikhailovsky, A.; Zasadzinski, J.A. Scalable routes to gold nanoshells with tunable sizes and response to near-infrared pulsed-laser irradiation. Small 2008, 4, 1183–1195, doi:10.1002/smll.200701290.
[21]  Rasch, M.R.; Sokolov, K.V.; Korgel, B.A. Limitations on the optical tunability of small diameter gold nanoshells. Langmuir 2009, 25, 11777–11785, doi:10.1021/la901249j.
[22]  Storti, B.; Elisei, F.; Abbruzzetti, S.; Viappiani, C.; Latterini, L. One-pot synthesis of gold nanoshells with high photon-to-heat conversion efficiency. J. Phys. Chem. C 2009, 113, 7516–7521.
[23]  Chithrani, B.D.; Chan, W.C.W. Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes. Nano Lett. 2007, 7, 1542–1550.
[24]  Morino, M.; Tsuzuki, T.; Ishikawa, Y.; Shirakami, T.; Yoshimura, M.; Kiyosuke, Y.I.; Matsunaga, K.; Yoshikumi, C.; Saijo, N. Specific expression of HSP27 in human tumour cell lines in vitro. In Vivo 1997, 11, 179–184.
[25]  Sreedhar, A.S.; Csermely, P. Heat shock proteins in the regulation of apoptosis: New strategies in tumour therapy—A comprehensive review. Pharmacol. Ther. 2004, 101, 227–257, doi:10.1016/j.pharmthera.2003.11.004.
[26]  Kamada, M.; So, A.; Muramaki, M.; Rocchi, P.; Beraldi, E.; Gleave, M. Hsp27 knockdown using nucleotide-based therapies inhibit tumour growth and enhance chemotherapy in human bladder cancer cells. Mol. Cancer Ther. 2007, 6, 299–308, doi:10.1158/1535-7163.MCT-06-0417.
[27]  Rocchi, P.; Jugpal, P.; So, A.; Sinneman, S.; Ettinger, S.; Fazli, L.; Nelson, C.; Gleave, M. Small interference RNA targeting heat-shock protein 27 inhibits the growth of prostatic cell lines and induces apoptosis via caspase-3 activation in vitro. BJU Int. 2006, 98, 1082–1089, doi:10.1111/j.1464-410X.2006.06425.x.
[28]  Cornford, P.A.; Dodson, A.R.; Parsons, K.F.; Desmond, A.D.; Woolfenden, A.; Fordham, M.; Neoptolemos, J.P.; Ke, Y.Q.; Foster, C.S. Heat shock protein expression independently predicts clinical outcome in prostate cancer. Cancer Res. 2000, 60, 7099–7105.
[29]  Dozmorov, M.G.; Hurst, R.E.; Culkin, D.J.; Kropp, B.P.; Frank, M.B.; Osban, J.; Penning, T.M.; Lin, H.K. Unique patterns of molecular profiling between human prostate cancer LNCaP and PC-3 cells. Prostate 2009, 69, 1077–1090.
[30]  Tate, A.; Isotani, S.; Bradley, M.J.; Sikes, R.A.; Davis, R.; Chung, L.W.K.; Edlund, M. Met-independent hepatocyte growth factor-mediated regulation of cell adhesion in human prostate cancer cells. BMC Cancer 2006, 6, 197–212, doi:10.1186/1471-2407-6-197.
[31]  Gleave, M.; Miyake, H.; Chi, K. Beyond simple castration: targeting the molecular basis of treatment resistance in advanced prostate cancer. Cancer Chemother. Pharmacol. 2005, 56, S47–S57, doi:10.1007/s00280-005-0098-0.
[32]  Liu, Y.L.; Franzen, S. Factors determining the efficacy of nuclear delivery of antisense oligonucleotides by gold nanoparticles. Bioconjug. Chem. 2008, 19, 1009–1016, doi:10.1021/bc700421u.
[33]  Bilanges, B.; Stokoe, D. Direct comparison of the specificity of gene silencing using antisense oligonucleotides and RNAi. Biochem. J. 2005, 388, 573–583, doi:10.1042/BJ20041956.
[34]  Gabai, V.L.; Budagova, K.R.; Sherman, M.Y. Increased expression of the major heat shock protein Hsp72 in human prostate carcinoma cells is dispensable for their viability but confers resistance to a variety of anticancer agents. Oncogene 2005, 24, 3328–3338, doi:10.1038/sj.onc.1208495.
[35]  So, A.; Rocchi, P.; Gleave, M. Antisense oligonucleotide therapy in the management of bladder cancer. Curr. Opin. Urol. 2005, 15, 320–327, doi:10.1097/01.mou.0000175572.46986.2c.
[36]  Hadchity, E.; Aloy, M.T.; Paulin, C.; Armandy, E.; Watkin, E.; Rousson, R.; Gleave, M.; Chapet, O.; Rodriguez-Lafrasse, C. Heat shock protein 27 as a new therapeutic target for radiation sensitization of head and neck squamous cell carcinoma. Mol. Ther. 2009, 17, 1387–1394, doi:10.1038/mt.2009.90.
[37]  Rossi, A.; Ciafre, S.; Balsamo, M.; Pierimarchi, P.; Santoro, M.G. Targeting the heat shock factor 1 by RNA interference: A potent tool to enhance hyperthermochemotherapy efficacy in cervical cancer. Cancer Res. 2006, 66, 7678–7685, doi:10.1158/0008-5472.CAN-05-4282.
[38]  Huang, H.C.; Barua, S.; Kay, D.B.; Rege, K. Simultaneous enhancement of photothermal stability and gene delivery efficacy of gold nanorods using polyelectrolytes. ACS Nano 2009, 3, 2941–2952, doi:10.1021/nn900947a.
[39]  Jain, P.K.; Ei-Sayed, M.A. Surface plasmon resonance sensitivity of metal nanostructures: Physical basis and universal scaling in metal nanoshells. J. Phys. Chem. C 2007, 111, 17451–17454, doi:10.1021/jp0773177.
[40]  Preston, T.C.; Signorell, R. Preparation and optical properties of metallodielectric core-shell-corona particles. J. Phys. Chem. C 2008, 112, 17844–17848.
[41]  Zhai, Y.M.; Zhai, J.F.; Wang, Y.L.; Guo, S.J.; Ren, W.; Dong, S.J. Fabrication of iron oxide core/gold shell submicrometer spheres with nanoscale surface roughness for efficient surface-enhanced raman scattering. J. Phys. Chem. C 2009, 113, 7009–7014.
[42]  Perez-Juste, J.; Pastoriza-Santos, I.; Liz-Marzan, L.M.; Mulvaney, P. Gold nanorods: Synthesis, characterization and applications. Coord. Chem. Rev. 2005, 249, 1870–1901.
[43]  Alberts, B. Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002.
[44]  Chithrani, B.D.; Ghazani, A.A.; Chan, W.C.W. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 2006, 6, 662–668, doi:10.1021/nl052396o.
[45]  Arnida, A.; Malugin, A.; Ghandehari, H. Cellular uptake and toxicity of gold nanoparticles in prostate cancer cells: A comparative study of rods and spheres. J. Appl. Toxicol. 2010, 30, 212–217.
[46]  Niidome, T.; Akiyama, Y.; Yamagata, M.; Kawano, T.; Mori, T.; Niidome, Y.; Katayama, Y. Poly(ethylene glycol)-modified gold nanorods as a photothermal nanodevice for hyperthermia. J. Biomater. Sci. 2009, 20, 1203–1215.
[47]  Hamblin, M.R.; Miller, J.L.; Rizvi, I.; Loew, H.G.; Hasan, T. Pegylation of charged polymer-photosensitiser conjugates: Effects on photodynamic efficacy. Br. J. Cancer 2003, 89, 937–943.
[48]  Calderwood, S.K.; Asea, A. Targeting HSP70-induced thermotolerance for design of thermal sensitizers. Int. J. Hyperth. 2002, 18, 597–608, doi:10.1080/0265673021000019666.
[49]  Stern, J.M.; Cadeddu, J.A. Emerging use of nanoparticles for the therapeutic ablation of urologic malignancies. Urol. Oncol.-Semin. Orig. Investig. 2008, 26, 93–96.
[50]  Cheng, F.Y.; Chen, C.T.; Yeh, C.S. Comparative efficiencies of photothermal destruction of malignant cells using antibody-coated silica@Au nanoshells, hollow Au/Ag nanospheres and Au nanorods. Nanotechnology 2009, 20, 425104–425113, doi:10.1088/0957-4484/20/42/425104.
[51]  Takahashi, H.; Niidome, T.; Nariai, A.; Niidome, Y.; Yamada, S. Photothermal reshaping of gold nanorods prevents further cell death. Nanotechnology 2006, 17, 4431–4435, doi:10.1088/0957-4484/17/17/024.
[52]  Stauffer, P.R.; Goldberg, S.N. Introduction: Thermal ablation therapy. Int. J. Hyperth. 2004, 20, 671–677, doi:10.1080/02656730400007220.
[53]  Stober, W.; Fink, A.; Bohn, E. Controlled growth of monodisperse silica spheres in micron size range. J. Colloid Interface Sci. 1968, 26, 62–69.
[54]  Sun, Y.Y.; Yan, F.; Yang, W.W.; Zhao, S.A.; Yang, W.S.; Sun, C.Q. Effect of silica nanoparticles with different sizes on the catalytic activity of glucose oxidase. Anal. Bioanal. Chem. 2007, 387, 1565–1572, doi:10.1007/s00216-006-1013-1.
[55]  Duff, D.G.; Baiker, A.; Edwards, P.P. A new hydrosol of gold clusters 1. Formation and particle size variation. Langmuir 1993, 9, 2301–2309, doi:10.1021/la00033a010.
[56]  Oldenburg, S.J.; Averitt, R.D.; Westcott, S.L.; Halas, N.J. Nanoengineering of optical resonances. Chem. Phys. Lett. 1998, 288, 243–247, doi:10.1016/S0009-2614(98)00277-2.
[57]  Hirsch, L.R.; Lee, R.; Halas, N.J.; West, J.L. Whole-Blood Immunoassay Facilitated by Gold Nanoshell-Conjugate Antibodies. In Nanobiotechnology Protocols; Rosenthal, S.J., Wright, D.W., Eds.; Humana Press: Totowa, NJ, USA, 2005; pp. 101–111.
[58]  Nikoobakht, B.; El-Sayed, M.A. Preparation and growth mechanism of gold nanorods (NRs) using seed-mediated growth method. Chem. Mater. 2003, 15, 1957–1962, doi:10.1021/cm020732l.
[59]  Au, L.; Zheng, D.S.; Zhou, F.; Li, Z.Y.; Li, X.D.; Xia, Y.N. A quantitative study on the photothermal effect of immuno gold nanocages targeted to breast cancer cells. ACS Nano 2008, 2, 1645–1652.
[60]  Liu, Y.L.; Shipton, M.K.; Ryan, J.; Kaufman, E.D.; Franzen, S.; Feldheim, D.L. Synthesis, stability, and cellular internalization of gold nanoparticles containing mixed peptide-poly(ethylene glycol) monolayers. Anal. Chem. 2007, 79, 2221–2229.
[61]  Weiss, A.; Preston, T.C.; Popov, J.; Li, Q.F.; Wu, S.; Chou, K.C.; Burt, H.M.; Bally, M.B.; Signorell, R. Selective recognition of rituximab-functionalized gold nanoparticles by lymphoma cells studied with 3D Imaging. J. Phys. Chem. C 2009, 113, 20252–20258.
[62]  Leung, J.P. Photothermal Therapy of Prostate Cancer using Gold Nanoparticles. M.Sc. Thesis, University of British Columbia, Vancouver, Canada, November 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133