Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Mini-Review Article

A Review on Cancer Therapy Based on the Photothermal Effect of Gold Nanorod

Author(s): Weizhen Xu, Qinlu Lin, Yueqin Yin, Dong Xu*, Xiaohui Huang, Bucheng Xu and Guangwei Wang*

Volume 25, Issue 46, 2019

Page: [4836 - 4847] Pages: 12

DOI: 10.2174/1381612825666191216150052

Price: $65

Abstract

Background: Cancer causes millions of deaths and huge economic losses every year. The currently practiced methods for cancer therapy have many defects, such as side effects, low curate rate, and discomfort for patients.

Objective: Herein, we summarize the applications of gold nanorods (AuNRs) in cancer therapy based on their photothermal effect-the conversion of light into local heat under irradiation.

Methods: The recent advances in the synthesis and regulation of AuNRs, and facile surface functionalization further facilitate their use in cancer treatment. For cancer therapy, AuNRs need to be modified or coated with biocompatible molecules (e.g. polyethylene glycol) and materials (e.g. silicon) to reduce the cytotoxicity and increase their biocompatibility, stability, and retention time in the bloodstream. The accumulation of AuNRs in cancerous cells and tissues is due to the high leakage in tumors or the specific interaction between the cell surface and functional molecules on AuNRs such as antibodies, aptamers, and receptors.

Results: AuNRs are employed not only as therapeutics to ablate tumors solely based on the heat produced under laser that could denature protein and activate the apoptotic pathway, but also as synergistic therapies combined with photodynamic therapy, chemotherapy, and gene therapy to kill cancer more efficiently. More importantly, other materials like TiO2, graphene oxide, and silicon, etc. are incorporated on the AuNR surface for multimodal cancer treatment with high drug loadings and improved cancer-killing efficiency. To highlight their applications in cancer treatment, examples of therapeutic effects both in vitro and in vivo are presented.

Conclusion: AuNRs have potential applications for clinical cancer therapy.

Keywords: Photothermal effect, gold nanorod, cancer therapy, photothermal therapy, synergistic therapy, cancer-killing efficiency.

[1]
Lee EYHP, Muller WJ. Oncogenes and tumor suppressor genes. Cold Spring Harb Perspect Biol 2010; 2(10) a003236
[http://dx.doi.org/10.1007/978-3-642-85076-9_4]
[2]
De Guzman R, Malik M. Global cancer burden and natural disasters: a focus on Asia’s vulnerability, resilience building, and impact on cancer care. J Glob Oncol 2019; 5: 1-8.
[http://dx.doi.org/10.1200/JGO.19.00037]
[3]
Kapiteijn E, Marijnen CA, Nagtegaal ID, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med 2001; 345: 638-46.
[http://dx.doi.org/10.1056/NEJMoa010580]
[4]
Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 783-92.
[http://dx.doi.org/10.1056/NEJM200103153441101]
[5]
Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006; 354: 567-78.
[http://dx.doi.org/10.1056/NEJMoa053422]
[6]
Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med 2001; 345: 725-30.
[http://dx.doi.org/10.1056/NEJMoa010187]
[7]
Zhang W, Guo Z, Huang D, Liu Z, Guo X, Zhong H. Synergistic effect of chemo-photothermal therapy using PEGylated graphene oxide. Biomaterials 2011; 32: 8555-61.
[http://dx.doi.org/10.1016/j.biomaterials.2011.07.071]
[8]
Muller-Runkel R, Kalokhe U, Schreiber G. Radiation therapy of unusually large breasts: using higher energy photons with or w/o boluS for all or part of the treatment course. Int J Radiat Oncol Biol Phys 1991; 21: 227.
[http://dx.doi.org/10.1016/0360-3016(91)90635-H]
[9]
Adams DM, Brus L, Chidsey CED, et al. Charge transfer on the nanoscale: current status. J Phys Chem B 2003; 107: 6668-97.
[http://dx.doi.org/10.1021/jp0268462]
[10]
Elghanian R, Storhoff JJ, Mucic RC, Letsinger RL, Mirkin CA. Selective colorimetric detection of polynucleotides based on the distance-dependent optical properties of gold nanoparticles. Sci Sports 1997; 277: 1078-81.
[11]
Mazhuga AG, Volkova NV, Manzhelii EA, Beloglazkina EK, Zyk NV, Zefirov NS. New nanohybrid material based on gold nanoparticles and 1,4-bis(terpyridine-4′-yl)benzene. Nanotechnol Russ 2012; 7: 149-51.
[http://dx.doi.org/10.1134/S1995078012020139]
[12]
Thomas L, Lionti F, Ballou R, Gatteschi D, Sessoli R, Barbara B. Macroscopic quantum tunnelling of magnetization in a single crystal of nanomagnets. Nature 1996; 383: 145-7.
[http://dx.doi.org/10.1038/383145a0]
[13]
Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer therapy and diagnosis. Adv Drug Deliv Rev 2002; 54: 631-51.
[http://dx.doi.org/10.1016/S0169-409X(02)00044-3]
[14]
Huang X, El-Sayed IH, Qian W, El-Sayed MA. Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J Am Chem Soc 2006; 128: 2115-20.
[http://dx.doi.org/10.1021/ja057254a]
[15]
Sun X, Liu Z, Welsher K, et al. Nano-graphene oxide for cellular imaging and drug delivery. Nano Res 2008; 1: 203-12.
[http://dx.doi.org/10.1007/s12274-008-8021-8]
[16]
Chen H, Shao L, Li Q, Wang J. Gold nanorods and their plasmonic properties. Chem Soc Rev 2013; 42: 2679-724.
[http://dx.doi.org/10.1039/C2CS35367A]
[17]
Pérez-Juste J, Pastoriza-Santos I, Liz-Marzán LM, Mulvaney P. Gold nanorods: synthesis, characterization and applications. Coord Chem Rev 2005; 249: 1870-901.
[http://dx.doi.org/10.1016/j.ccr.2005.01.030]
[18]
Vigderman L, Khanal BP, Zubarev ER. Functional gold nanorods: synthesis, self assembly, and sensing applications. Adv Mater 2012; 24: 4811-41.
[http://dx.doi.org/10.1002/adma.201201690]
[19]
Sharma V, Park K, Srinivasarao M. Colloidal dispersion of gold nanorods: historical background, optical properties, seed-mediated synthesis, shape separation and self-assembly. Mater Sci Eng Rep 2009; 65: 1-38.
[http://dx.doi.org/10.1016/j.mser.2009.02.002]
[20]
Orendorff CJ, Gole A, Sau TK, Murphy CJ. Surface-enhanced Raman spectroscopy of self-assembled monolayers: sandwich architecture and nanoparticle shape dependence. Anal Chem 2005; 77: 3261-6.
[http://dx.doi.org/10.1021/ac048176x]
[21]
Tong L, Wei Q, Wei A, Cheng J-X. Gold nanorods as contrast agents for biological imaging: optical properties, surface conjugation and photothermal effects. Photochem Photobiol 2009; 85: 21-32.
[http://dx.doi.org/10.1111/j.1751-1097.2008.00507.x]
[22]
Wang H, Huff TB, Zweifel DA, et al. In vitro and in vivo two-photon luminescence imaging of single gold nanorods. Proc Natl Acad Sci USA 2005; 102: 15752-6.
[http://dx.doi.org/10.1073/pnas.0504892102]
[23]
Du Y, Jiang Q, Beziere N, et al. DNA-nanostructure-gold-nanorod hybrids for enhanced in vivo optoacoustic imaging and photothermal therapy. Adv Mater 2016; 28: 10000-7.
[http://dx.doi.org/10.1002/adma.201601710]
[24]
Yamashita T, Forgues M, Wang W, et al. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res 2008; 68: 1451-61.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-6013]
[25]
Zheng G, Patolsky F, Cui Y, Wang WU, Lieber CM. Multiplexed electrical detection of cancer markers with nanowire sensor arrays. Nat Biotechnol 2005; 23: 1294-301.
[http://dx.doi.org/10.1038/nbt1138]
[26]
Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008; 105: 10513-8.
[http://dx.doi.org/10.1073/pnas.0804549105]
[27]
Li J-L, Gu M. Surface plasmonic gold nanorods for enhanced two-photon microscopic imaging and apoptosis induction of cancer cells. Biomaterials 2010; 31: 9492-8.
[http://dx.doi.org/10.1016/j.biomaterials.2010.08.068]
[28]
Vigderman L, Zubarev ER. High-yield synthesis of gold nanorods with longitudinal SPR peak greater than 1200 nm using hydroquinone as a reducing agent. Chem Mater 2013; 25: 1450-7.
[http://dx.doi.org/10.1021/cm303661d]
[29]
Weissleder R. A clearer vision for in vivo imaging. Nat Biotechnol 2001; 19: 316-7.
[http://dx.doi.org/10.1038/86684]
[30]
Oyelere AK, Chen PC, Huang X, El-Sayed IH, El-Sayed MA. Peptide-conjugated gold nanorods for nuclear targeting. Bioconjug Chem 2007; 18: 1490-7.
[http://dx.doi.org/10.1021/bc070132i]
[31]
Chen R, Zheng X, Qian H, Wang X, Wang J, Jiang X. Combined near-IR photothermal therapy and chemotherapy using gold-nanorod/chitosan hybrid nanospheres to enhance the antitumor effect. Biomater Sci 2013; 1: 285-93.
[http://dx.doi.org/10.1039/C2BM00138A]
[32]
Chevallier P, Walter A, Garofalo A, et al. Tailored biological retention and efficient clearance of pegylated ultra-small MnO nanoparticles as positive MRI contrast agents for molecular imaging. J Mater Chem B Mater Biol Med 2014; 2: 1779-90.
[http://dx.doi.org/10.1039/C3TB21634A]
[33]
Lee H, Lee JH, Kim J, et al. Hyaluronate-gold nanorod/DR5 antibody complex for noninvasive theranosis of skin cancer. ACS Appl Mater Interfaces 2016; 8: 32202-10.
[http://dx.doi.org/10.1021/acsami.6b11319]
[34]
Hong Y, Lee E, Choi J, et al. Gold nanorod-mediated photothermal modulation for localized ablation of cancer cells. J Nanomater 2012; 2012 825060
[http://dx.doi.org/10.1155/2012/825060]
[35]
Liao J, Li W, Peng J, et al. Combined cancer photothermal-chemotherapy based on doxorubicin/gold nanorod-loaded polymersomes. Theranostics 2015; 5: 345-56.
[http://dx.doi.org/10.7150/thno.10731]
[36]
O’Neal DP, Hirsch LR, Halas NJ, Payne JD, West JL. Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett 2004; 209: 171-6.
[http://dx.doi.org/10.1016/j.canlet.2004.02.004]
[37]
Sultan R. Tumour ablation by laser in general surgery. Lasers Med Sci 1990; 5: 185-93.
[http://dx.doi.org/10.1007/BF02031380]
[38]
Link S, El-Sayed MA. Shape and size dependence of radiative, non-radiative and photothermal properties of gold nanocrystals. Int Rev Phys Chem 2000; 19: 409-53.
[http://dx.doi.org/10.1080/01442350050034180]
[39]
Abbasian M, Mahmoodzadeh F, Amirshaghaghi OA, Salehi R. Chemo-photothermal therapy of cancer cells using gold nanorods-cored stimuli-responsive triblock copolymer. New J Chem 2017; 41: 12777-88.
[http://dx.doi.org/10.1039/C7NJ02504A]
[40]
Ni Q, Teng Z, Dang M, et al. Gold nanorod embedded large-pore mesoporous organosilica nanospheres for gene and photothermal cooperative therapy of triple negative breast cancer. Nanoscale 2017; 9: 1466-74.
[http://dx.doi.org/10.1039/C6NR07598C]
[41]
Wang J, Tang HY, Yang WL, Chen JY. Aluminum phthalocyanine and gold nanorod conjugates: the combination of photodynamic therapy and photothermal therapy to kill cancer cells. J Porphyr Phthalocyanines 2012; 16: 802-8.
[http://dx.doi.org/10.1142/S108842461250099X]
[42]
Cao J, Sun T, Grattan KTV. Gold nanorod-based localized surface plasmon resonance biosensors: a review. Sens Actuators B Chem 2014; 195: 332-51.
[http://dx.doi.org/10.1016/j.snb.2014.01.056]
[43]
Ma Z, Xia H, Liu Y, Liu B, Chen W, Zhao Y. Applications of gold nanorods in biomedical imaging and related fields. Chin Sci Bull 2013; 58: 2530-6.
[http://dx.doi.org/10.1007/s11434-013-5720-7]
[44]
Wang X, Shao M, Zhang S, Liu X. Biomedical applications of gold nanorod-based multifunctional nano-carriers. J Nanopart Res 2013; 15: 1892.
[http://dx.doi.org/10.1007/s11051-013-1892-y]
[45]
Wang J, Zhang HZ, Li RS, Huang CZ. Localized surface plasmon resonance of gold nanorods and assemblies in the view of biomedical analysis. TrAC Trends Analyt Chem 2016; 80: 429-43.
[http://dx.doi.org/10.1016/j.trac.2016.03.015]
[46]
Haine AT, Niidome T. Gold nanorods as nanodevices for bioimaging, photothermal therapeutics, and drug delivery. Chem Pharm Bull (Tokyo) 2017; 65: 625-8.
[http://dx.doi.org/10.1248/cpb.c17-00102]
[47]
Li X-F, Chen C-Y, Zhao Y-H, Yuan X-Y. Surface modification of gold nanorods and their applications in combination of cancer diagnosis and therapy. Prog Biochem Biophys 2014; 41: 739-48.
[48]
Jana NR, Gearheart LA, Murphy CJ. Seed-mediated growth approach for shape-controlled synthesis of spheroidal and rod-like gold nanoparticles using a surfactant template. Adv Mater 2001; 13: 1389-93.
[http://dx.doi.org/10.1002/1521-4095(200109)13:18<1389:AID-ADMA1389>3.0.CO;2-F]
[49]
Chang S-S, Shih C-W, Chen C-D, Lai W-C, Wang CC. The shape transition of gold nanorods. Langmuir 1999; 15: 701-9.
[http://dx.doi.org/10.1021/la980929l]
[50]
Martin CR. Nanomaterials: a membrane-based synthetic approach. Science 1994; 266: 1961-6.
[http://dx.doi.org/10.1126/science.266.5193.1961]
[51]
Evans P, Hendren W, Atkinson R, et al. Growth and properties of gold and nickel nanorods in thin film alumina. Nanotechnology 2006; 17: 5746.
[http://dx.doi.org/10.1088/0957-4484/17/23/006]
[52]
Huang C-J, Chiu P-H, Wang Y-H, Yang C-F. Synthesis of the gold nanodumbbells by electrochemical method. J Colloid Interface Sci 2006; 303: 430-6.
[http://dx.doi.org/10.1016/j.jcis.2006.07.073]
[53]
Yu Y-Y, Chang S-S, Lee C-L, Wang CC. Gold nanorods: electrochemical synthesis and optical properties. J Phys Chem B 1997; 101: 6661-4.
[http://dx.doi.org/10.1021/jp971656q]
[54]
Kabashin AV, Evans P, Pastkovsky S, et al. Plasmonic nanorod metamaterials for biosensing. Nat Mater 2009; 8: 867-71.
[http://dx.doi.org/10.1038/nmat2546]
[55]
Huang X, Neretina S, El Sayed MA. Gold nanorods: from synthesis and properties to biological and biomedical applications. Adv Mater 2009; 21: 4880-910.
[http://dx.doi.org/10.1002/adma.200802789]
[56]
Ye X, Jin L, Caglayan H, et al. Improved size-tunable synthesis of monodisperse gold nanorods through the use of aromatic additives. ACS Nano 2012; 6: 2804-17.
[http://dx.doi.org/10.1021/nn300315j]
[57]
Vigderman L, Zubarev ER. High-yield synthesis ofgold nanorods with longitudinal spr peak greater than 1200 nm using hydroquinone as a reducing agent. Chem Mater 2013; 25: 1450-7.
[http://dx.doi.org/10.1021/cm303661d]
[58]
Xu D, Mao J, He Y, Yeung ES. Size-tunable synthesis of high-quality gold nanorods under basic conditions by using H2O2 as the reducing agent. J Mater Chem C Mater Opt Electron Devices 2014; 2: 4989-96.
[http://dx.doi.org/10.1039/c4tc00483c]
[59]
Liopo A, Wang S, Derry PJ, Oraevsky AA, Zubarev ER. Seedless synthesis of gold nanorods using dopamine as a reducing agent. RSC Advances 2015; 5: 91587-93.
[http://dx.doi.org/10.1039/C5RA19817H]
[60]
Park K, Hsiao M-S, Yi Y-J, et al. Highly concentrated seed-mediated synthesis of monodispersed gold nanorods. ACS Appl Mater Interfaces 2017; 9: 26363-71.
[http://dx.doi.org/10.1021/acsami.7b08003]
[61]
Chang H-H, Murphy CJ. Mini gold nanorods with tunable plasmonic peaks beyond 1000 nm. Chem Mater 2018; 30: 1427-35.
[http://dx.doi.org/10.1021/acs.chemmater.7b05310]
[62]
Qiu Y, Liu Y, Wang L, et al. Surface chemistry and aspect ratio mediated cellular uptake of Au nanorods. Biomaterials 2010; 31: 7606-19.
[http://dx.doi.org/10.1016/j.biomaterials.2010.06.051]
[63]
Lu L, Xia Y. Enzymatic reaction modulated gold nanorod end-to-end self-assembly for ultrahigh sensitively colorimetric sensing of cholinesterase and organophosphate pesticides in human blood. Anal Chem 2015; 87: 8584-91.
[http://dx.doi.org/10.1021/acs.analchem.5b02516]
[64]
Orendorff CJ, Alam TM, Sasaki DY, Bunker BC, Voigt JA. Phospholipid-gold nanorod composites. ACS Nano 2009; 3: 971-83.
[http://dx.doi.org/10.1021/nn900037k]
[65]
Kou X, Zhang S, Tsung C-K, et al. Growth of gold nanorods and bipyramids using CTEAB surfactant. J Phys Chem B 2006; 110: 16377-83.
[http://dx.doi.org/10.1021/jp0639086]
[66]
He J, Unser S, Bruzas I, et al. The facile removal of CTAB from the surface of gold nanorods. Colloids Surf B Biointerfaces 2018; 163: 140-5.
[http://dx.doi.org/10.1016/j.colsurfb.2017.12.019]
[67]
Niidome T, Yamagata M, Okamoto Y, et al. PEG-modified gold nanorods with a stealth character for in vivo applications. J Control Release 2006; 114: 343-7.
[http://dx.doi.org/10.1016/j.jconrel.2006.06.017]
[68]
Akiyama Y, Mori T, Katayama Y, Niidome T. The effects of PEG grafting level and injection dose on gold nanorod biodistribution in the tumor-bearing mice. J Control Release 2009; 139: 81-4.
[http://dx.doi.org/10.1016/j.jconrel.2009.06.006]
[69]
Li Y, Jin J, Wang D, et al. Coordination-responsive drug release inside gold nanorod@metal-organic framework core-shell nanostructures for near-infrared-induced synergistic chemo-photothermal therapy. Nano Res 2018; 11: 3294-305.
[http://dx.doi.org/10.1007/s12274-017-1874-y]
[70]
Xu W, Qian J, Hou G, et al. A dual-targeted hyaluronic acid-gold nanorod platform with triple-stimuli responsiveness for photodynamic/photothermal therapy of breast cancer. Acta Biomater 2019; 83: 400-13.
[http://dx.doi.org/10.1016/j.actbio.2018.11.026]
[71]
Zhang C, Zhang F, Wang W, et al. Chitosan coated gold nanorod chelating gadolinium for MRI-visible photothermal therapy of cancer. RSC Advances 2016; 6: 111337-44.
[http://dx.doi.org/10.1039/C6RA23769J]
[72]
Cobley CM, Jingyi C, Eun Chul C, Wang LV, Younan X. Gold nanostructures: a class of multifunctional materials for biomedical applications. Chem Soc Rev 2010; 40: 44-56.
[http://dx.doi.org/10.1039/B821763G]
[73]
Alkilany AM, Thompson LB, Boulos SP, Sisco PN, Murphy CJ. Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Adv Drug Deliv Rev 2012; 64: 190-9.
[http://dx.doi.org/10.1016/j.addr.2011.03.005]
[74]
Park H, Lee S, Chen L, et al. SERS imaging of HER2-overexpressed MCF7 cells using antibody-conjugated gold nanorods. Phys Chem Chem Phys 2009; 11: 7444-9.
[http://dx.doi.org/10.1039/b904592a]
[75]
Bonoiu AC, Bergey EJ, Ding H, et al. Gold nanorod-siRNA induces efficient in vivo gene silencing in the rat hippocampus. Nanomed 2011; 6: 617-30.
[http://dx.doi.org/10.2217/nnm.11.20]
[76]
Xu L, Liu Y, Chen Z, et al. Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. Nano Lett 2012; 12: 2003-12.
[http://dx.doi.org/10.1021/nl300027p]
[77]
Liu Z, Wang L, Zhang L, et al. Metabolic characteristics of 16HBE and A549 cells exposed to different surface modified gold nanorods. Adv Healthc Mater 2016; 5: 2363-75.
[http://dx.doi.org/10.1002/adhm.201600164]
[78]
Chen Y-S, Frey W, Kim S, et al. Enhanced thermal stability of silica-coated gold nanorods for photoacoustic imaging and image-guided therapy. Opt Express 2010; 18: 8867-77.
[http://dx.doi.org/10.1364/OE.18.008867]
[79]
Jokerst JV, Thangaraj M, Kempen PJ, Sinclair R, Gambhir SS. Photoacoustic imaging of mesenchymal stem cells in living mice via silica-coated gold nanorods. ACS Nano 2012; 6: 5920-30.
[http://dx.doi.org/10.1021/nn302042y]
[80]
Zhong Y, Wang C, Wang X, et al. Gold nanorod-cored micelles as a remotely-controllable near IR-triggered drug release system for effective killing of drug resistant cancer cells. J Control Release 2013; 172: 86-7.
[http://dx.doi.org/10.1016/j.jconrel.2013.08.176]
[81]
Mirza AZ, Shamshad H. Fabrication and characterization of doxorubicin functionalized PSS coated gold nanorod. Arab J Chem 2019; 12: 146-50.
[http://dx.doi.org/10.1016/j.arabjc.2014.08.009]
[82]
Chen M, Qiu P, He X, et al. The adenine DNA self-assembly of pH- and near-infrared-responsive gold nanorod vehicles for the chemothermal treatment of cancer cells. J Mater Chem B Mater Biol Med 2014; 2: 3204-13.
[http://dx.doi.org/10.1039/c4tb00103f]
[83]
Manivasagan P, Hoang G, Moorthy MS, et al. Chitosan/fucoidan multilayer coating of gold nanorods as highly efficient near-infrared photothermal agents for cancer therapy. Carbohydr Polym 2019; 211: 360-9.
[http://dx.doi.org/10.1016/j.carbpol.2019.01.010]
[84]
Wang L, Li D, Hao Y, et al. Gold nanorod-based poly(lactic-co-glycolic acid) with manganese dioxide core-shell structured multifunctional nanoplatform for cancer theranostic applications. Int J Nanomedicine 2017; 12: 3059-74.
[http://dx.doi.org/10.2147/IJN.S128844]
[85]
Zhu F, Tan G, Jiang Y, Yu Z, Ren F. Rational design of multi-stimuli-responsive gold nanorod-curcumin conjugates for chemo-photothermal synergistic cancer therapy. Biomater Sci 2018; 6: 2905-17.
[http://dx.doi.org/10.1039/C8BM00691A]
[86]
Chuang C-C, Cheng C-C, Chen P-Y, et al. Gold nanorod-encapsulated biodegradable polymeric matrix for combined photothermal and chemo-cancer therapy. Int J Nanomedicine 2019; 14: 181-93.
[http://dx.doi.org/10.2147/IJN.S177851]
[87]
Durr NJ, Larson T, Smith DK, Korgel BA, Sokolov K, Ben-Yakar A. Two-photon luminescence imaging of cancer cells using molecularly targeted gold nanorods. Nano Lett 2007; 7: 941-5.
[http://dx.doi.org/10.1021/nl062962v]
[88]
Van Du N, Min H-K, Kim C-S, Han J, Park J-O, Choi E. Folate receptor-targeted liposomal nanocomplex for effective synergistic photothermal-chemotherapy of breast cancer in vivo. Colloids Surf B Biointerfaces 2019; 173: 539-48.
[http://dx.doi.org/10.1016/j.colsurfb.2018.10.013]
[89]
Zhang Y, Zhan X, Xiong J, et al. Temperature-dependent cell death patterns induced by functionalized gold nanoparticle photothermal therapy in melanoma cells. Sci Rep 2018; 8: 8720.
[http://dx.doi.org/10.1038/s41598-018-26978-1]
[90]
Masood R, Roy I, Zu S, et al. Gold nanorod-sphingosine kinase siRNA nanocomplexes: a novel therapeutic tool for potent radiosensitization of head and neck cancer. Integr Biol 2012; 4: 132-41.
[http://dx.doi.org/10.1039/C1IB00060H]
[91]
Chen C-L, Kuo L-R, Chang C-L, et al. In situ real-time investigation of cancer cell photothermolysis mediated by excited gold nanorod surface plasmons. Biomaterials 2010; 31: 4104-12.
[http://dx.doi.org/10.1016/j.biomaterials.2010.01.140]
[92]
Wu Y, Ali MRK, Dong B, et al. Gold nanorod photothermal therapy alters cell junctions and actin network in inhibiting cancer cell collective migration. ACS Nano 2018; 12: 9279-90.
[http://dx.doi.org/10.1021/acsnano.8b04128]
[93]
Dickerson EB, Dreaden EC, Huang X, et al. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett 2008; 269: 57-66.
[http://dx.doi.org/10.1016/j.canlet.2008.04.026]
[94]
Mooney R, Schena E, Saccomandi P, Zhumkhawala A, Aboody K, Berlin JM. Gold nanorod-mediated near-infrared laser ablation: in vivo experiments on mice and theoretical analysis at different settings. Int J Hyperthermia 2017; 33: 150-9.
[http://dx.doi.org/10.1080/02656736.2016.1230682]
[95]
Song J, Yang X, Jacobson O, et al. Ultrasmall gold nanorod vesicles with enhanced tumor accumulation and fast excretion from the body for cancer therapy. Adv Mater 2015; 27: 4910-7.
[http://dx.doi.org/10.1002/adma.201502486]
[96]
Zhang M, Kim HS, Jin T, Woo J, Piao YJ, Moon WK. Near-infrared photothermal therapy using anti-EGFR-gold nanorod conjugates for triple negative breast cancer. Oncotarget 2017; 8: 86566-75.
[http://dx.doi.org/10.18632/oncotarget.21243]
[97]
Zhang Y, He J, Wang Y, et al. Photothermal therapy with AuNRs and EGFRmAb-AuNRs inhibits subcutaneous transplantable hypopharyngeal tumors in nude mice. Int J Oncol 2018; 53: 2647-58.
[http://dx.doi.org/10.3892/ijo.2018.4559]
[98]
Bar H, Yacoby I, Benhar I. Killing cancer cells by targeted drug-carrying phage nanomedicines. BMC Biotechnol 2008; 8: 37-7.
[http://dx.doi.org/10.1186/1472-6750-8-37]
[99]
Zeng J-Y, Zhang M-K, Peng M-Y, Gong D, Zhang X-Z. Porphyrinic metal-organic frameworks coated gold nanorods as a versatile nanoplatform for combined photodynamic/photothermal/chemotherapy of tumor. Adv Funct Mater 2018; 28 1705451
[http://dx.doi.org/10.1002/adfm.201705451]
[100]
Ko H, Son S, Bae S, Kim J-H, Yi G-R, Park JH. Near-infrared light-triggered thermochemotherapy of cancer using a polymer-gold nanorod conjugate. Nanotechnology 2016; 27(14)175102
[http://dx.doi.org/10.1088/0957-4484/27/17/175102]
[101]
Zhong Y, Wang C, Cheng L, Meng F, Zhong Z, Liu Z. Gold nanorod-cored biodegradable micelles as a robust and remotely controllable doxorubicin release system for potent inhibition of drug-sensitive and - resistant cancer cells. Biomacromolecules 2013; 14: 2411-9.
[http://dx.doi.org/10.1021/bm400530d]
[102]
Tu T-Y, Yang S-J, Tsai M-H, et al. Dual-triggered drug-release vehicles for synergistic cancer therapy. Colloids Surf B Biointerfaces 2019; 173: 788-97.
[http://dx.doi.org/10.1016/j.colsurfb.2018.10.043]
[103]
Zhu F, Tan G, Zhong Y, et al. Smart nanoplatform for sequential drug release and enhanced chemo-thermal effect of dual drug loaded gold nanorod vesicles for cancer therapy. J Nanobiotechnology 2019; 17: 44.
[http://dx.doi.org/10.1186/s12951-019-0473-3]
[104]
Hao Y, Dong M, Zhang T, et al. Novel approach of using near-infrared responsive pegylated gold nanorod coated poly(l-lactide) microneedles to enhance the antitumor efficiency of docetaxel-loaded MPEG-PDLLA micelles for treating an A431 tumor. ACS Appl Mater Interfaces 2017; 9: 15317-27.
[http://dx.doi.org/10.1021/acsami.7b03604]
[105]
Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer 2006; 6: 535-45.
[http://dx.doi.org/10.1038/nrc1894]
[106]
Choi J, Lee S-E, Park J-S, Kim SY. Gold nanorod-photosensitizer conjugates with glutathione-sensitive linkages for synergistic cancer photodynamic/photothermal therapy. Biotechnol Bioeng 2018; 115: 1340-54.
[http://dx.doi.org/10.1002/bit.26536]
[107]
Zhang P, Wang C, Zhao J, et al. Near infrared-guided smart nanocarriers for microRNA-controlled release of doxorubicin/siRNA with intracellular ATP as fuel. ACS Nano 2016; 10: 3637-47.
[http://dx.doi.org/10.1021/acsnano.5b08145]
[108]
Wang B-K, Yu X-F, Wang J-H, et al. Gold-nanorods-siRNA nanoplex for improved photothermal therapy by gene silencing. Biomaterials 2016; 78: 27-39.
[http://dx.doi.org/10.1016/j.biomaterials.2015.11.025]
[109]
Shen J, Kim H-C, Mu C, et al. Multifunctional gold nanorods for siRNA gene silencing and photothermal therapy. Adv Healthc Mater 2014; 3: 1629-37.
[http://dx.doi.org/10.1002/adhm.201400103]
[110]
Yi Y, Wang H, Wang X, Liu Q, Ye M, Tan W. A smart, photocontrollable drug release nanosystem for multifunctional synergistic cancer therapy. ACS Appl Mater Interfaces 2017; 9: 5847-54.
[http://dx.doi.org/10.1021/acsami.6b15414]
[111]
Leng C, Zhang X, Xu F, et al. Engineering gold nanorod-copper sulfide heterostructures with enhanced photothermal conversion efficiency and photostability. Small 2018; 14 1703077
[http://dx.doi.org/10.1002/smll.201703077]
[112]
Li Y, Pan G, Liu Q, et al. Coupling resonances of surface plasmon in gold nanorod/copper chalcogenide core-shell nanostructures and their enhanced photothermal effect. ChemPhysChem 2018; 19: 1852-8.
[http://dx.doi.org/10.1002/cphc.201701338]
[113]
Cui X, Cheng W, Han X. Lipid bilayer modified gold nanorod@mesoporous silica nanoparticles for controlled drug delivery triggered by near-infrared light. J Mater Chem B Mater Biol Med 2018; 6: 8078-84.
[http://dx.doi.org/10.1039/C8TB01891J]
[114]
Lee J, Lee YH, Jeong CB, Choi JS, Chang KS, Yoon M. Gold nanorods-conjugated TiO2 nanoclusters for the synergistic combination of phototherapeutic treatments of cancer cells. J Nanobiotechnology 2018; 16: 104.
[http://dx.doi.org/10.1186/s12951-018-0432-4]
[115]
Sun B, Wu J, Cui S, et al. In situ synthesis of graphene oxide/gold nanorods theranostic hybrids for efficient tumor computed tomography imaging and photothermal therapy. Nano Res 2017; 10: 37-48.
[http://dx.doi.org/10.1007/s12274-016-1264-x]
[116]
Assali A, Razzazan S, Akhavan O, Mottaghitalab F, Adeli M, Atyabi F. The bio-interface between functionalized Au NR@GO nanoplatforms with protein corona and their impact on delivery and release system. Colloids Surf B Biointerfaces 2019; 173: 891-8.
[http://dx.doi.org/10.1016/j.colsurfb.2018.10.042]
[117]
Jia Q, Ge J, Liu W, et al. Gold nanorod@silica-carbon dots as multifunctional phototheranostics for fluorescence and photoacoustic imaging-guided synergistic photodynamic/photothermal therapy. Nanoscale 2016; 8: 13067-77.
[http://dx.doi.org/10.1039/C6NR03459D]
[118]
Jiang Q, Shi Y, Zhang Q, et al. A self-assembled DNA origami-gold nanorod complex for cancer theranostics. Small 2015; 11: 5134-41.
[http://dx.doi.org/10.1002/smll.201501266]
[119]
Wang C, Xu C, Xu L, et al. A novel core-shell structured upconversion nanorod as a multimodal bioimaging and photothermal ablation agent for cancer theranostics. J Mater Chem B Mater Biol Med 2018; 6: 2597-607.
[http://dx.doi.org/10.1039/C7TB02842C]
[120]
Liu H, Liu T, Li L, et al. Size dependent cellular uptake, in vivo fate and light-heat conversion efficiency of gold nanoshells on silica nanorattles. Nanoscale 2012; 4: 3523-9.
[http://dx.doi.org/10.1039/c2nr30396e]
[121]
Jia H, Fang C, Zhu X-M, Ruan Q, Wang Y-XJ, Wang J. Synthesis of absorption-dominant small gold nanorods and their plasmonic properties. Langmuir 2015; 31: 7418-26.
[http://dx.doi.org/10.1021/acs.langmuir.5b01444]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy