Micro-CT enables microlocalisation and quantification of Her2-targeted gold nanoparticles within tumour regions.

OBJECTIVES Gold nanoparticles are of interest as potential in vivo diagnostic and therapeutic agents, as X-ray contrast agents, drug delivery vehicles and radiation enhancers. The aim of this study was to quantitatively determine their targeting and microlocalisation in mouse tumour models after intravenous injection by using micro-CT. METHODS Gold nanoparticles (15 nm) were coated with polyethylene glycol and covalently coupled to anti-Her2 antibodies (Herceptin). In vitro, conjugates incubated with Her2+ (BT-474) and Her2- (MCF7) human breast cancer cells showed specific targeted binding with a Her2+ to Her2- gold ratio of 39.4±2.7:1. Nude mice, simultaneously bearing subcutaneous Her2+ and Her2- human breast tumours in opposite thighs were prepared. Gold nanoparticles alone, conjugated to Herceptin or to a non-specific antibody were compared. After intravenous injection of the gold nanoparticles, gold concentrations were determined by atomic absorption spectroscopy. Microlocalisation of gold was carried out by calibrated micro-CT, giving both the radiodensities and gold concentrations in tumour and non-tumour tissue. RESULTS All gold nanoparticle constructs showed accumulation, predominantly at tumour peripheries. However, the Herceptin-gold nanoparticles showed the best specific uptake in their periphery (15.8±1.7% injected dose per gram), 1.6-fold higher than Her2- tumours and 22-fold higher than surrounding muscle. Imaging readily enabled detection of small, 1.5 mm-thick tumours. CONCLUSION In this pre-clinical study, antibody-targeted 15 nm gold nanoparticles showed preferential uptake in cognate tumours, but even untargeted gold nanoparticles enhanced the visibility of tumour peripheries and enabled detection of millimetre-sized tumours. Micro-CT enabled quantification within various regions of a tumour.

[1]  C. Barraclough,et al.  Effects of estradiol and progesterone on plasma gonadotropins, prolactin, and LHRH in specific brain areas of ovariectomized rats. , 1981, Biology of reproduction.

[2]  R M Albrecht,et al.  Gastrointestinal persorption and tissue distribution of differently sized colloidal gold nanoparticles. , 2001, Journal of pharmaceutical sciences.

[3]  Noriaki Ohuchi,et al.  In vivo single molecular imaging and sentinel node navigation by nanotechnology for molecular targeting drug-delivery systems and tailor-made medicine , 2008, Breast cancer.

[4]  Zhuang Liu,et al.  Selective probing and imaging of cells with single walled carbon nanotubes as near-infrared fluorescent molecules. , 2008, Nano letters.

[5]  J. H. Hubbell,et al.  Tables of X-Ray Mass Attenuation Coefficients and Mass Energy-Absorption Coefficients 1 keV to 20 MeV for Elements Z = 1 to 92 and 48 Additional Substances of Dosimetric Interest , 1995 .

[6]  M Geso,et al.  Gold nanoparticles: a new X-ray contrast agent. , 2007, The British journal of radiology.

[7]  Kazunori Kataoka,et al.  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. , 2005, Biomacromolecules.

[8]  Sang Hyun Cho,et al.  Estimation of tumour dose enhancement due to gold nanoparticles during typical radiation treatments: a preliminary Monte Carlo study , 2005, Physics in medicine and biology.

[9]  P. Wise,et al.  Printed in U.S.A. Copyright © 2001 by The Endocrine Society Neuroprotective Effects of Estradiol in Middle-Aged Female Rats* , 2000 .

[10]  D. Slamon,et al.  Antibody to HER-2/neu receptor blocks DNA repair after cisplatin in human breast and ovarian cancer cells. , 1994, Oncogene.

[11]  May D. Wang,et al.  In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags , 2008, Nature Biotechnology.

[12]  R. Offord,et al.  Biokinetics of a F(ab')3 iodine-131 labeled antigen binding construct (Mab 35) directed against CEA in patients with colorectal carcinoma. , 2001, Cancer biotherapy & radiopharmaceuticals.

[13]  Robert Jones,et al.  A Phase I Trial of Humanized Monoclonal Antibody A33 in Patients with Colorectal Carcinoma: Biodistribution, Pharmacokinetics, and Quantitative Tumor Uptake , 2005, Clinical Cancer Research.

[14]  S. Gambhir,et al.  Small-Animal PET Imaging of Human Epidermal Growth Factor Receptor Type 2 Expression with Site-Specific 18F-Labeled Protein Scaffold Molecules , 2008, Journal of Nuclear Medicine.

[15]  Z. Bhujwalla,et al.  PAMAM dendrimer‐based contrast agents for MR imaging of Her‐2/neu receptors by a three‐step pretargeting approach , 2008, Magnetic resonance in medicine.

[16]  S. Ametamey,et al.  In vivo Evaluation of 177Lu- and 67/64Cu-Labeled Recombinant Fragments of Antibody chCE7 for Radioimmunotherapy and PET Imaging of L1-CAM-Positive Tumors , 2005, Clinical Cancer Research.

[17]  M J Paulus,et al.  High resolution X-ray computed tomography: an emerging tool for small animal cancer research. , 2000, Neoplasia.

[18]  Deborah E Leckband,et al.  HER-2-mediated endocytosis of magnetic nanospheres and the implications in cell targeting and particle magnetization. , 2008, Biomaterials.

[19]  J. Hainfeld,et al.  Radiotherapy enhancement with gold nanoparticles , 2008, The Journal of pharmacy and pharmacology.

[20]  Michael J Sailor,et al.  Computationally guided photothermal tumor therapy using long-circulating gold nanorod antennas. , 2009, Cancer research.

[21]  V. Chernomordik,et al.  Affibody Molecules for In vivo Characterization of HER2-Positive Tumors by Near-Infrared Imaging , 2008, Clinical Cancer Research.

[22]  C. McCollough,et al.  Dual-energy CT iodine-subtraction virtual unenhanced technique to detect urinary stones in an iodine-filled collecting system: a phantom study. , 2008, AJR. American journal of roentgenology.

[23]  Tristan Barrett,et al.  In vivo molecular imaging to diagnose and subtype tumors through receptor-targeted optically labeled monoclonal antibodies. , 2007, Neoplasia.

[24]  R. Reilly,et al.  Imaging of HER2/neu-positive BT-474 human breast cancer xenografts in athymic mice using (111)In-trastuzumab (Herceptin) Fab fragments. , 2005, Nuclear medicine and biology.

[25]  Valery V Tuchin,et al.  Circulation and distribution of gold nanoparticles and induced alterations of tissue morphology at intravenous particle delivery , 2009, Journal of biophotonics.

[26]  B. Yeh,et al.  Renal cyst pseudoenhancement at multidetector CT: what are the effects of number of detectors and peak tube voltage? , 2008, Radiology.

[27]  Raoul Kopelman,et al.  Targeted gold nanoparticles enable molecular CT imaging of cancer. , 2008, Nano letters.

[28]  S. Cohen,et al.  Direct visualization of the binding and internalization of a ferritin conjugate of epidermal growth factor in human carcinoma cells A-431 , 1979, The Journal of cell biology.

[29]  Keishiro Tomoda,et al.  Biodistribution of colloidal gold nanoparticles after intravenous administration: effect of particle size. , 2008, Colloids and surfaces. B, Biointerfaces.

[30]  S. Nie,et al.  Nanotechnology applications in cancer. , 2007, Annual review of biomedical engineering.

[31]  H. Dvorak,et al.  Identification and characterization of the blood vessels of solid tumors that are leaky to circulating macromolecules. , 1988, The American journal of pathology.

[32]  P. Cuatrecasas,et al.  Adsorbents for affinity chromatography. Use of N-hydroxysuccinimide esters of agarose. , 1972, Biochemistry.

[33]  Hiroshi Maeda,et al.  Early Phase Tumor Accumulation of Macromolecules: A Great Difference in Clearance Rate between Tumor and Normal Tissues , 1998, Japanese journal of cancer research : Gann.

[34]  Sibaprasad Bhattacharyya,et al.  Synthesis and evaluation of near-infrared (NIR) dye-herceptin conjugates as photoacoustic computed tomography (PCT) probes for HER2 expression in breast cancer. , 2008, Bioconjugate chemistry.

[35]  M. Brechbiel,et al.  Novel bimodal bifunctional ligands for radioimmunotherapy and targeted MRI. , 2008, Bioconjugate chemistry.

[36]  R. Johnstone,et al.  Comparison of in vitro cell binding characteristics of four monoclonal antibodies and their individual tumor localization properties in mice. , 1990, Cancer research.

[37]  U. Nielsen,et al.  Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models. , 2006, Cancer research.

[38]  Kwon-Ha Yoon,et al.  Colloidal Gold Nanoparticles as a Blood-Pool Contrast Agent for X-ray Computed Tomography in Mice , 2007, Investigative radiology.

[39]  J. West,et al.  Near-infrared resonant nanoshells for combined optical imaging and photothermal cancer therapy. , 2007, Nano letters.

[40]  G. Frens Controlled Nucleation for the Regulation of the Particle Size in Monodisperse Gold Suspensions , 1973 .

[41]  Hillyer,et al.  Correlative Instrumental Neutron Activation Analysis, Light Microscopy, Transmission Electron Microscopy, and X-ray Microanalysis for Qualitative and Quantitative Detection of Colloidal Gold Spheres in Biological Specimens , 1998, Microscopy and Microanalysis.

[42]  R. Paxton,et al.  Preoperative imaging of colorectal carcinoma with 111In-labeled anticarcinoembryonic antigen monoclonal antibody. , 1986, Cancer research.

[43]  Stephen J McMahon,et al.  Radiotherapy in the presence of contrast agents: a general figure of merit and its application to gold nanoparticles , 2008, Physics in medicine and biology.

[44]  Dong Liang,et al.  Influence of anchoring ligands and particle size on the colloidal stability and in vivo biodistribution of polyethylene glycol-coated gold nanoparticles in tumor-xenografted mice. , 2009, Biomaterials.

[45]  Takuro Niidome,et al.  PEG-modified gold nanorods with a stealth character for in vivo applications. , 2006, Journal of controlled release : official journal of the Controlled Release Society.

[46]  J. Hainfeld,et al.  The use of gold nanoparticles to enhance radiotherapy in mice. , 2004, Physics in medicine and biology.

[47]  Lawrence Tamarkin,et al.  Colloidal Gold: A Novel Nanoparticle Vector for Tumor Directed Drug Delivery , 2004, Drug delivery.