AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

EGFR Antibody Conjugated Bimetallic Au@Ag Nanorods for Enhanced SERS-Based Tumor Boundary Identification, Targeted Photoacoustic Imaging and Photothermal Therapy

Shouhui Chen1,2Chenchen Bao1Chunlei Zhang1Yao Yang1Kan Wang1Bhaskara V. Chikkaveeraiah2,3Zhihua Wang1,2Xinglu Huang2Fei Pan1Kun Wang1Xiao Zhi1Jian Ni1( )Jesus M. de la Fuente1,5( )Jie Tian4
Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China
Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
Microfabrication and Microfluidics Unit, Biomedical Engineering and Physical Science Shared Resource, National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
Intelligent Medical Research Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
Instituto de Nanociencia de Aragon (INA), Universidad de Zaragoza, Zaragoza, 50018, Spain
Show Author Information

Abstract

The development of multifunctional nanoprobes for simultaneous targeted imaging, tumor boundary identification and photothermal therapy of gastric cancer has become a great challenge. Herein, EGFR monoclonal antibody-conjugated Au@Ag nanorods decorated with DTNB nanoprobes (5,5'-Dithiobis-(2-nitrobenzoic acid)) (EGFR-Au@Ag NR nanotags) were prepared and characterized. Their biocompatibility was analyzed by MTT assay. In vitro studies show that EGFR-Au@Ag NR nanotags own good biocompatibility and capability of targeting and entering into gastric cancer MGC803 cells, silver nano-film layer on the surface of gold nanorods remarkably enhance surface-enhanced rama spectra (SERS) signal, enhanced photoacoustic imaging efficacy and photothermal conversion efficiency of gold nanorods. In vivo studies show that prepared nanotags could target actively gastric cancer cells at 2h post-injection, and distributed in the tumor site, exhibited enhanced SERS signals to display clearly tumor boundary, enhanced photoacoustic imaging to display clearly tumor boundary. Under 1w/cm2 laser irradiation, tumor growth was remarkably inhibited by local photothermal therapy. In conclusion, high performance EGFR-antibody conjugated Au @ Ag nanorod-DTNB nanoprobes exhibit great potential in applications such as targeted photoacoustic imaging, simultaneous tumor boundary identification and selective photothermal therapy of gastric cancer in the near future.

Electronic Supplementary Material

Download File(s)
nbe-8-4-315_ESM.pdf (281.8 KB)

References

[1]

J. Ferlay, H.R. Shin, F. Bray, et al., Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer, 2010, 127: 2893-2917.

[2]

R. Siegel, D. Naishadham, and A. Jemal, Cancer statistics, 2013. CA Cancer J Clin, 2013, 63: 11–30.

[3]

T. Takahashi, Y. Saikawa, and Y. Kitagawa, Gastric cancer: current status of diagnosis and treatment. Cancers (Basel), 2013, 5: 48-63.

[4]

I. Proserpio, S. Rausei, S. Barzaghi, et al., Multimodal treatment of gastric cancer. World Journal of Gastrointestinal Surgery, 2014, 6: 55-58.

[5]

D. Zhang, D. Fan, New insights into the mechanisms of gastric cancer multidrug resistance and future perspectives. Future Oncol, 2010, 6: 527-537.

[6]

D. Cui, L. Zhang, X.J. Yan, et al., A microarray-based gastric carcinoma prewarning system. World J Gastroenterol, 2005, 11: 1273-1282.

[7]

C. Bao, J. Conde, F. Pan, et al., Gold nanoprisms as a hybrid in vivo cancer theranostic platform for in situ photoacoustic imaging, angiography, and localized hyperthermia. Nano Research, 2016, 9: 1043-1056.

[8]

M.F. Clarke, J.E. Dick, P.B. Dirks, et al., Cancer stem cells-perspectives on current status and future directions: Aacr workshop on cancer stem cells. Cancer Res, 2006, 66: 9339-9344.

[9]

C. Zhang, C. Li, F. He, et al., Identification of cd44+cd24+ gastric cancer stem cells. J Cancer Res Clin Oncol, 2011, 137: 1679-1686.

[10]

K. Wang, J. Ruan, Q. Qian, et al., BRCAA1 monoclonal antibody conjugated fluorescent magnetic nanoparticles for in vivo targeted magnetofluorescent imaging of gastric cancer. J Nanobiotechnol, 20111, 9: 23.

[11]

C. Wang, C.C. Bao, S.J. Liang, et al., HAI-178 antibody-conjugated fluorescent magnetic nanoparticles for targeted imaging and simultaneous therapy of gastric cancer. Nanoscale Res. Lett, 2014, 9: 274.

[12]

J. Ruan, H. Song, Q.R. Qian, et al., HER2 monoclonal antibody conjugated RNase-A-associated CdTe quantum dots for targeted imaging and therapy of gastric cancer. Biomaterials, 2012, 33: 7093-7102.

[13]

M. He, P. Huang, C.L. Zhang, et al., Dual phase-controlled synthesis of uniform lanthanide-doped NaGdF4 upconversion nanocrystals via an OA/ionic liquid two-phase system for in vivo dual-modality imaging. Advanced Functional Materials, 2011, 21: 4470 - 4477.

[14]

Z.M. Li, P. Huang, X.J. Zhang, et al., RGD-conjugated dendrimer-modified gold nanorods for in vivo tumor targeting and photothermal therapy. Molecular Pharmaceutics, 2010, 7: 94-104.

[15]

P. Huang, J. Lin, X.S. Wang, et al., Light-triggered theranostics based on photosensitizer-conjugated carbon dots for simultaneous enhanced-fluorescence imaging and photodynamic therapy. Adv. Mater, 2012, 24: 5104–5110.

[16]

Z.J. Zhou, C.L. Zhang, Q.R. Qian, et al., Folic acid-conjugated silica capped gold nanoclusters for targeted fluorescence/X-ray computed tomography imaging. Journal of Nanobiotechnology, 2013, 11: 17.

[17]

C.L. Zhang, Z.J. Zhou, Q.R. Qian, et al., Glutathione-capped fluorescent gold nanoclusters for dual-modal fluorescence/X-ray computed tomography imaging. J. Mater. Chem. B, 2013, 1: 5045-5053.

[18]

S.J. Liang, C. Li, C.L. Zhang, et al., CD44v6 Monoclonal Antibody-Conjugated Gold Nanostars for Targeted Photoacoustic Imaging and Plasmonic Photothermal Therapy of Gastric Cancer Stem-like Cells. Theranostics, 2015, 5: 970-984.

[19]

D.X. Cui, C.L. Zhang, B. Liu, et al., Regression of gastric cancer by systemic injection of RNA nanoparticles carrying both ligand and siRNA. Sci Rep, 2015: 10726.

[20]

P. Huang, C. Xu, J. Lin, et al., Folic acid-conjugated graphene oxide loaded with photosensitizers for targeting photodynamic therapy. Theranostics,. 2011, 1: 240-250.

[21]

J.P. Zhang, Y. Song, C.L. Zhang, et al., Circulating MiR-16-5p and MiR-19b-3p as two novel potential biomarkers to indicate progression of gastric cancer. Theranostics, 2015, 5, 733-745.

[22]

Y.X. Zhang, G. Gao, H.J. Liu, et al.,.Identification of volatile biomarkers of gastric cancer cells and ultrasensitive electrochemical detection based on sensing interface of au-ag alloy coated MWCNTs. Theranostics, 2014, 4: 154-162.

[23]

C. Wang, C.C. Bao, S.J. Liang, et al., RGD-conjugated silica-coated gold nanorods on the surface of carbon nanotubes for targeted photoacoustic imaging of gastric cancer. Nanoscale Res. Lett, 2014, 9: 264.

[24]

P. Huang, L. Bao, C.L. Zhang, et al., Folic acid-conjugated silica-modified gold nanorods for X-ray/CT imaging-guided dual-mode radiation and photo-thermal therapy. Biomaterials, 2011, 32: 9796-9809.

[25]

Y. Liu, M. Yang, J. Zhang, et al., Human induced pluripotent stem cells for tumor targeted delivery of gold nanorods and enhanced photothermal therapy. ACS Nano, 2016, 10: 2375-2385.

[26]

S.H. Chen, P. Huang, Z.H. Wang, et al., Self-assembly of gold nanoparticles to silver microspheres as highly efficient 3D SERS substrates. Nanoscale Res. Lett, 2013, 8: 168.

[27]

X.S. Wang, D.P. Yang, P. Huang, et al., Hierarchically assembled Au microsphers and ser urchin-like architectures: formation mechanism and SERS study. Nanoscale, 2012, 4: 7766.

[28]

L. Wang, T. Guo, Q. Lu, et al., Sea-urchin-like Au nanocluster with surface-enhanced Raman scattering in detecting epidermal growth factor receptor (EGFR) mutation status of malignant pleural effusion. ACS Applied Materials & Interface, 2015, 7: 359-369.

[29]

S.H. Chen, D.B. Liu, Z.H. Wang, et al., Picomolar detection of mercuric ions by means of gold silver core-shell nanorod. Nanoscale, 2013, 5: 6731-6735

[30]

C.C. Bao, N. Beziere, P. del Pino, et al., Gold nanoprisms as optoacoustic signal nanoamplifiers for in vivo bioimaging of gastrointestinal cancers. Small, 2013, 9: 68-74.

[31]

C.L. Zhang, C. Li, Y.L. Liu, et al., Gold nanoclusters-based nanoprobes for simultaneous fluorescence imaging and targeted photodynamic therapy with superior penetration and retention behavior in tumors. Advanced Fun. Mater, 2015, 25: 1314-1325.

[32]

C. Bao, J. Conde, E. Polo, et al., A promising road with challenges: where gold nanoparticles are in translational research? Nanomedicine, 2014, 9: 2353-2370.

[33]

C.J. Orendorff, T.K. Sau, and C.J. Murphy, Shape-dependent plasmon-resonant gold nanoparticles. Small, 2006, 2: 636-639.

[34]

X. Huang, I.H. El-Sayed, W. Qian, et al., Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. Journal of the American Chemical Society, 2006, 128: 2115-2120.

[35]

L.L. Feng, G. Gao, P. Huang, et al., Optical proerties and catalytic activity of bimetallic gold -silver nanoparticles. Nano Biomed Eng, 2010, 2: 258-267.

[36]

C.J. Orendorff, T.K. Sau, C.J. Murphy, Shape-dependent plasmon-resonant gold nanoparticles. Small, 2006, 2(5): 636-639.

[37]

B. Pelaz, P. del Pino, Synthesis and applications of gold nanoparticles. Frontiers of Nanoscience, 2012, 4: 3-33.

[38]

J. Conde, C.C. Bao, D.X. Cui, et al., Antibody-drug gold nanoantennas with Raman spectroscopic fingerprints for in vivo tumor theranostics J. Controlled Release, 2014, 183: 87-93.

[39]

C.C. Bao, J. Conde, J. Curtin, et al., Bioresponsive antisense DNA gold nanobeacons as a hybrid in vivo theranostics platform for the inhibition of cancer cells and metastasis. Sci Rep, 2015, 5: 12197.

[40]

B.F. Pan, D.X. Cui, C. Ozkan, et al., DNA-templated ordered array of gold nanorods in one and two dimesnions. J. Phys. Chem. C, 2007, 111 (34): 12572–12576.

[41]

G.V. Hartland, G. Schatz, Virtual issue: plasmon resonances-a physical chemistry perspective. The Journal of Physical Chemistry C, 2011, 115(31): 15121-15123.

[42]

S.M. Leto, F. Sassi, I. Catalano, et al., Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2-amplified gastrointestinal carcinomas. Clin Cancer Res, 2015, 21(24): 5519-5531.

[43]

J. Han, J. Zhang, M. Yang, et al., Glucose-functionalized Au nanoprisms for optoacoustic imaging and near-infrared photothermal therapy. Nanoscale, 2016, 8: 492-499.

[44]

C. Li, S. Liang, C. Zhang, et al., Allogenic dendritic cell and tumor cell fused vaccine for targeted imaging and enhanced immunotherapeutic efficacy of gastric cancer. Biomaterials, 2015, 54: 177-187.

[45]

J. Conde, C.C. Bao, Y.Q. Tan,et al., Dual targeted immunotherapy via in vivo delivery of biohybrid RNAi-peptide nanoparticles to tumor-associated macrophages and cancer cells. Adv. Fun. Mater, 2015, 25: 4183-4194.

[46]

W. Lu, Q. Huang, G. Ku,et al., Photoacoustic imaging of living mouse brain vasculature using hollow gold nanospheres. Biomaterials, 2010, 31(9): 2617-2626.

[47]

M.A. Mackey, M.R. Ali, L.A. Austin, et al., The most effective gold nanorod size for plasmonic photothermal therapy: theory and in vitro experiments. The Journal of Physical Chemistry B, 2014, 118(5): 1319-1326.

[48]

Y. Li, T. Wen, R. Zhao, et al., Localized electric field of plasmonic nanoplatform enhanced photodynamic tumor therapy. ACS Nano, 2014, 8(11): 11529-11542.

[49]

A.M. Alkilany, L.B. Thompson, S.P. Boulos, et al., Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Advanced Drug Delivery Reviews, 2012, 64(2): 190-199.

[50]

N.J. Durr, T. Larson, D.K. Smith, et al., Two-photon luminescence imaging of cancer cells using molecularly targeted gold nanorods. Nano Letters, 2007, 7(4): 941-945.

Nano Biomedicine and Engineering
Pages 315-328
Cite this article:
Chen S, Bao C, Zhang C, et al. EGFR Antibody Conjugated Bimetallic Au@Ag Nanorods for Enhanced SERS-Based Tumor Boundary Identification, Targeted Photoacoustic Imaging and Photothermal Therapy. Nano Biomedicine and Engineering, 2016, 8(4): 315-328. https://doi.org/10.5101/nbe.v8i4.p315-328

400

Views

6

Downloads

14

Crossref

18

Scopus

Altmetrics

Received: 22 December 2016
Accepted: 27 December 2016
Published: 30 December 2016
© 2016 Shouhui Chen, Chenchen Bao, Chunlei Zhang, Yao Yang, Kan Wang, Bhaskara V. Chikkaveeraiah, Zhihua Wang, Xinglu Huang, Fei Pan, Kun Wang, Xiao Zhi, Jian Ni, Jesus M. de la Fuente, and Jie Tian.

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return