Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nucl Med Biol ; 42(2): 85-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25441255

ABSTRACT

INTRODUCTION: (18)F-2-deoxy-2-fluoro-d-glucose ((18)F-FDG) positron emission tomography (PET) has been used for imaging human cancers for several decades. Despite its extensive use, (18)F-FDG PET imaging has limitations in the tumor findings. The goal of this study was to investigate the potential of a PPAR-γ agonist pioglitazone (PIO) to distinguish tumors and inflammatory lesions in (18)F-FDG PET imaging. METHODS: Studies of cellular uptake of (18)F-FDG and Western blot were performed in macrophage (RAW264.7) and three tumor cell lines (A549, KB, and MDA-MB-231) after treatment with PIO. In vivo microPET/CT imaging and biodistribution were performed in animal models. RESULTS: The uptake of (18)F-FDG in the macrophages was decreased and uptake of (18)F-FDG in the tumor cells was increased when these cells were treated with PIO. Western blot showed that the expression of Glut1 was reduced by treatment of PIO in the macrophage cells, whereas the expression of Glut1 in the tumor cells was increased. In vivo PET/CT imaging revealed that (18)F-FDG uptake (%ID/g) in the tumors was enhanced from 4.05±1.46 to 5.28±1.92 for A549, from 3.9±0.5 to 4.9±0.2 for KB, and from 9.14±0.86 to 13.48±2.07 for MDA-MB-231 tumors after treatment with PIO. Unlike tumors, the RAW264.7 xenograft model showed the reduced (18)F-FDG uptake in the inflammatory lesion from 11.74±1.19 to 6.50±1.47. The results of biodistribution also showed that (18)F-FDG uptake in the tumors were increased after treatment of PIO. However, the uptake of inflammation lesions was reduced. CONCLUSIONS: In this study, we demonstrated the effect of a PPAR-γ agonist PIO on (18)F-FDG uptake in tumors and inflammation in vitro and in vivo. PIO has potential to differentiate tumors and inflammatory lesions on (18)F-FDG PET imaging.


Subject(s)
Fluorodeoxyglucose F18 , Neoplasms/diagnostic imaging , PPAR gamma/agonists , Positron-Emission Tomography , Thiazolidinediones/pharmacology , Anilides/pharmacology , Animals , Biological Transport/drug effects , Cell Line, Tumor , Diagnosis, Differential , Female , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/antagonists & inhibitors , Humans , Inflammation/diagnostic imaging , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Neoplasms/metabolism , PPAR gamma/antagonists & inhibitors , Pioglitazone , Tissue Distribution/drug effects
2.
J Nucl Med ; 54(11): 1974-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24050935

ABSTRACT

UNLABELLED: A new approach to the surface engineering of superparamagnetic iron oxide nanoparticles (SPIONs) may encourage their development for clinical use. In this study, we demonstrated that nonpolymeric surface modification of SPIONs has the potential to be an advanced biocompatible contrast agent for biomedical applications, including diagnostic imaging in vivo. METHODS: Adenosine triphosphate (ATP), which is an innate biomaterial derived from the body, was coated onto the surface of SPIONs. An in vivo degradation study of ATP-coated SPIONs (ATP@SPIONs) was performed for 28 d. To diminish phagocytosis, ATP@SPIONs were surface-modified with gluconic acid. We next studied the ability of the SPIONs to serve as a specific targeted contrast agent after conjugation of cMet-binding peptide. The SPIONs were conjugated with Cy5.5 and labeled with (125)I for multimodality imaging. In vivo and in vitro tumor-targeted binding studies were performed on U87MG cells or a U87MG tumor model using animal SPECT/CT, an optical imaging system, and a 1.5-T clinical MR scanner. RESULTS: ATP@SPIONs showed rapid degradation in vivo and in vitro, compared with ferumoxides. ATP@SPIONs modified with gluconic acid reduced phagocytic uptake, showed improved biodistribution, and provided good targetability in vivo. The gluconic acid-conjugated ATP@SPIONs, when conjugated with cMet-binding peptide, were successfully visualized on the U87MG tumors implanted in mice via multimodality imaging. CONCLUSION: We suggest that ATP@SPIONs can be used as a multiplatform to target a region of interest in molecular imaging. When we consider the biocompatibility of contrast agents in vivo, ATP@SPIONs are superior to polymeric surface-modified SPIONs.


Subject(s)
Coated Materials, Biocompatible/metabolism , Ferric Compounds/chemistry , Molecular Imaging/methods , Nanoparticles/metabolism , Adenosine Triphosphate/chemistry , Animals , Cell Line, Tumor , Coated Materials, Biocompatible/chemistry , Gluconates/chemistry , Humans , Ligands , Magnets/chemistry , Mice , Multimodal Imaging , Nanoparticles/chemistry , Surface Properties
3.
J Liposome Res ; 23(4): 291-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23738810

ABSTRACT

The imaging of sentinel lymph nodes (SLN) has been researched for its role in assessing cancer progression and postsurgical lymphedema. Indocyanine green (ICG) is a near-infrared (NIR) optical dye that has been approved by the Food and Drug Administration. It is known that liposome-encapsulated ICG (LP-ICG) has improved stability and fluorescence signal compared with ICG. We designed mannosylated liposome-encapsulated ICG (M-LP-ICG) as an optical contrast agent for SLN. M-LP-ICG has a higher UV absorbance spectrum and fluorescence intensity than LP-ICG. The stability of M-LP-ICG measured in 50% fetal bovine serum solution by a dialysis method was better than that of LP-ICG. M-LP-ICG demonstrated a high uptake in RAW 264.7 macrophage cell because the density of mannose is high. There were differences between M-LP-ICG and glucosylated liposome-encapsulated ICG (G-LP-ICG), which are geometrical isomers. The result of an inhibition study of M-LP-ICG showed a statistically significant decrease in uptake in RAW 264.7 cells after either co-treatment or pre-treatment with D-(+)-mannose as an inhibitor. Results from an in vitro experiment demonstrated that M-LP-ICG was specifically taken up by macrophage cells through the mannose receptor on its surface. The time-series images acquired from a normal mouse model after subcutaneous injection showed that the signal from M-LP-ICG in SLN and other organs appeared early and disappeared quickly in comparison with signals from LP-ICG. Not only the sentinel but also the draining lymph nodes were observed partly in M-LP-ICG. M-LP-ICG appears to increase the specificity of uptake and retention in macrophages, making it a good candidate contrast agent for an optic imaging system for SLN and the lymphatic system.


Subject(s)
Indocyanine Green/administration & dosage , Liposomes , Lymph Nodes/pathology , Mannose/metabolism , Animals , Cattle , Cell Line , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence
4.
Nucl Med Biol ; 39(6): 805-12, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22421431

ABSTRACT

The epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor and plays an important role in carcinogenesis. In this study, the epidermal growth factor receptor binding peptide (EGBP) was identified using a phage display method and evaluated in U87MG cells in order to investigate the possibility to target the EGFR using an optical imaging system. Cyanine dye 5.5 (Cy5.5) was conjugated with EGBP-GGG-SC, EGBP-AOC-SC, and EGBP-AM2BA-SC. Cellular binding study of EGBP-Linker-Cy5.5 conjugates or (125)I-EGBP-Linker compounds was performed in U87MG cells. Optical imaging studies were performed in U87MG bearing mice. Three of seven clones from the 12-mer peptide library showed a specific binding affinity to rhEGFR, and they encoded the same 12 amino acid peptide sequence, FPMFNHWEQWPP. Confocal images show that the fluorescent signal of EGBP-Linker-Cy5.5 conjugates was decreased in the order: EGBP-AOC-Cy5.5≫EGBP-AM2BA-Cy5.5>EGBP-GGG-Cy5.5. EGBP-AOC-Cy5.5 appeared in cell cytoplasm and surface, and it was inhibited by free EGBP apparently. The cellular binding of EGBP-AOC-Cy5.5 exhibited a higher average radiance value than EGBP-GGG-Cy5.5 and EGBP-AM2BA-Cy5.5. Among various (125)I-EGBP-Linker compounds, EGBP-GGG showed a higher binding than other compounds. However, uptake of (125)I-EGBP-AOC was clearly inhibited by free EGBP in inhibition study. In an in vivo study, the fluorescent signal of EGBP-AOC-Cy5.5 treated mouse was mainly detected in the tumor and kidney. Among the three derivatives, EGBP-AOC-Cy5.5 was the optimized optical imaging agent for U87MG EGFR positive tumors in the animal model. This study demonstrated the EGBP-Linker-Cy5.5 conjugates may be useful as a potential EGFR target optical probe.


Subject(s)
Carbocyanines/chemistry , ErbB Receptors/metabolism , Fluorescent Dyes/chemistry , Oligopeptides/chemistry , Oligopeptides/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Optical Imaging , Peptide Library
5.
Int J Cancer ; 131(8): 1846-53, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22287125

ABSTRACT

The purpose of this study was to use a near-infrared (NIR) fluorescent cyclic His-Try-Gly-Phe peptide to characterize and image the expressions of matrix metalloproteinases (MMPs), which are correlated with cancer promotion, in an inflammation-induced colorectal cancer (ICRC) model. We explored the relationship between the development of colon cancer and the expression of MMPs at the same colonic sites in ICRC models. To develop ICRC models, mice were administered a single intraperitoneal dose (10 mg/kg) of azoxymethane (AOM) and exposed orally to 2% dextran sodium sulfate (DSS) for one week. MMP-2 expression and ß-catenin activation in colonic lesions were characterized by immunohistochemical (IHC) staining. After being treated with inducers for some time, cancerous lesions were found to express high ß-catenin and MMP-2. The profiles of MMP expression were correlated with ß-catenin activation in the colonic lesions. c(KAHWGFTLD)NH(2) (C6) peptide was prepared by standard Fmoc peptide synthesis to target MMPs. Molecular weight of Cy5.5-C6 was 1,954.78 g/mol (calculated MW = 1955.23 g/mol). The in vitro characterization of Cy5.5-C6 showed MMP binding specificity in a cell experiment. In vivo NIRF imaging showed high accumulation of Cy5.5-C6 in tumors with associated expression of MMP-2 in colonic lesions after intravenous injection. The MMP-2 specificity of Cy5.5-C6 was confirmed by successful inhibition of probe uptake in the tumor due to the presence of excess C6 peptide. The use of Cy5.5-C6 to target MMP-2 has the potential to be developed into an effective molecular imaging agent to monitor ICRC progress.


Subject(s)
Colonic Neoplasms/pathology , Diagnostic Imaging , Disease Models, Animal , Inflammation/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Azoxymethane/toxicity , Blotting, Western , Carbocyanines , Carcinogens/toxicity , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Dextran Sulfate/toxicity , Disease Progression , Immunoenzyme Techniques , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice , Mice, Inbred BALB C , Peptide Fragments/pharmacology , beta Catenin/metabolism
6.
Cancer Biother Radiopharm ; 27(2): 141-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22149589

ABSTRACT

Intact immunoglobulin G antibody has a relatively large molecule size of approximately 150 kDa that remains in the bloodstream for many weeks, which is a considerable disadvantage when it is used to carry radioactive materials for imaging. To lower background activity and increase the contrast of images, we investigated antivascular endothelial growth factor (VEGF) receptor 2 antibody (DC101) conjugated dextran for VEGF receptor 2 imaging in tumor xenografted mice. DTPA-conjugated aminodextran was synthesized, reacted with sulfo-LC-SPDP, and then reacted with DC101. The binding affinity of DTPA-dextran-DC101 to Flk-1 was measured. The gamma imaging and biodistributions of (99m)Tc-DTPA-dextran-DC101, (99m)Tc-DTPA-DC101, and (125)I-DC101 were studied in B16F10 melanoma xenografted mice. The dissociation values for DC101, DTPA-DC101, and DTPA-dextran-DC101 were 22.48, 3.05, and 14.74 pM, respectively. In gamma images, (99m)Tc-DTPA-dextran-DC101 showed weak liver uptake and rapid kidney elimination. In biodistribution results, the liver uptake of (99m)Tc-DTPA-dextran-DC101 was similar with that of (99m)Tc-DTPA-DC101 at each time point. However, the blood activity of (99m)Tc-DTPA-dextran-DC101 has shown significant differences, compared with (99m)Tc-DTPA-DC101 at all time points. The tumor accumulation of dextran-conjugated antibody was increased with time, whereas that of dextran nonconjugated antibody decreased. In particular, the pattern of tumor uptake of (99m)Tc-DTPA-dextran-DC101 was similar to that of (125)I-DC101, so this was thought to reflect the kinetics of DC101, unlike the nonconjugated form. The results of this study suggested that introduction of dextran moiety to make (99m)Tc-radiolabeled DC101 imaging agent could provide better images with the impaired background and the steady increasing binding to the receptor. However, further studies are necessary to improve clinical pharmacokinetics, such as enhancement of tumor uptake and impaired renal uptake.


Subject(s)
Antibodies, Monoclonal/chemistry , Dextrans/chemistry , Melanoma, Experimental/diagnostic imaging , Radiopharmaceuticals , Technetium , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Dextrans/pharmacokinetics , Female , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Pentetic Acid/chemistry , Pentetic Acid/pharmacokinetics , Pyridines/chemistry , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Succinates/chemistry , Technetium/chemistry , Technetium/pharmacokinetics , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Cancer Biother Radiopharm ; 26(6): 745-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22003972

ABSTRACT

Vascular endothelial growth factor receptor type 2 (VEGFR2)-targeted tumor treatment is an antiangiogenic therapeutic strategy. The human sodium iodide symporter (hNIS) gene is a useful reporter gene for tumor imaging and radiotherapy. In this study, we investigated the evaluation of therapeutic efficacy in hNIS gene-transfected tumor xenografts using a gamma imaging system after treatment with an anti-VEGFR2 antibody. Human breast cancer MDA-MB-231 cells transfected with the hNIS gene were injected subcutaneously into the right flanks of BALB/c nude mice. Therapy was initiated when the tumor volume reached approximately 130-180 mm(3). The animals were intravenously injected with 50, 100, or 150 µg of antibody every 3 days for 16 days. Gamma imaging was performed 1 and 2 weeks after the first injection to monitor the effects of tumor therapy. Mice were sacrificed 2 weeks after the first injection of antibody and the tumors were removed for CD31 staining and reverse transcription-polymerase chain reaction (RT-PCR) assay. All groups of mice that were treated with anti-hVEGFR2 antibody showed markedly reduced tumor growth compared to control mice. In vivo gamma imaging results showed that, at 1 week after the first injection of the anti-hVEGFR2 antibody, (125)I uptake of a tumor treated with 150 µg of antibody was 24.5% lower than that in the controls. At 2 weeks, (125)I uptake in the tumor treated with 150 µg of antibody was as low as 44.3% of that in the controls. CD31 staining and RT-PCR assays showed that blood vessel formation and expression of the hNIS gene were reduced with increased treatment doses. This study demonstrated the feasibility of molecular imaging and the therapeutic efficacy of developing therapeutic antibody anti-hVEGFR2 using a gamma imaging system in hNIS gene-transfected tumor xenograft mice.


Subject(s)
Antibodies/administration & dosage , Antibodies/immunology , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Molecular Imaging/methods , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Female , Genes, Reporter , Humans , Immunohistochemistry/methods , Iodine Radioisotopes/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Multimodal Imaging/methods , Neovascularization, Pathologic/genetics , Positron-Emission Tomography , Reproducibility of Results , Symporters/genetics , Tomography, X-Ray Computed , Transfection/methods , Tumor Burden/genetics , Xenograft Model Antitumor Assays/methods
8.
Nucl Med Biol ; 38(2): 287-94, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21315285

ABSTRACT

PURPOSE: The sodium iodide symporter (NIS) mediates active iodide uptake in lactating breast tissue, and when its levels are enhanced by all-trans retinoic acid (atRA), NIS has been proposed as a target for the imaging and radiotherapy of breast cancer. Importantly, the estrogen receptor α (ERα) is an important regulator of atRA induced NIS gene expression in breast cancer cells. In this study, we investigated the effect of an ER agonist (17ß-estradiol, E(2)) or antagonist [trans-hydroxytamoxifen (TOT) or raloxifene (RAL)] treatment on the regulation of NIS gene expression and iodide uptake in an ERα-positive breast cancer (MCF-7) model. METHODS: NIS functional activity was measured in vitro by (125)I uptake assay after incubation with E(2) (from 10(-15) to 10(-5) M), TOT (from 5×10(-8) to 5×10(-6) M), or RAL (from 5×10(-8) to 5×10(-6) M) in the presence or absence of atRA (10(-7) M). Under the same conditions, NIS mRNA expression was examined by reverse transcriptase polymerase chain reaction. Athymic mice with MCF-7 xenograft tumors were treated with atRA alone or atRA together with E(2) to evaluate the change of (125)I uptake in tumor tissues in vivo. RESULTS: In the iodide uptake study in cells, E(2), TOT, or RAL treatment alone did not stimulate (125)I uptake. However, when iodide uptake was stimulated by atRA, cotreatment with E(2), TOT or RAL decreased (125)I uptake in a concentration-dependent manner. The hormone effects on NIS mRNA expression levels in MCF-7 cells were similar. The results of the in vivo biodistribution study showed that (125)I uptake was reduced 50% in tumor tissues of mice treated with atRA/E(2) as compared to tumors treated only with atRA. CONCLUSION: Our results suggest that combination treatment of atRA and ER ligands could limit the functional activity of the NIS gene induced by atRA, thereby compromising its use as a target for diagnosis or radiotherapy in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Receptors, Estrogen/metabolism , Symporters/genetics , Animals , Biological Transport/drug effects , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Interactions , Estradiol/pharmacology , Female , Humans , Iodine Radioisotopes/metabolism , Iodine Radioisotopes/pharmacokinetics , Ligands , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tretinoin/pharmacology , Xenograft Model Antitumor Assays
9.
Nucl Med Biol ; 38(1): 93-101, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21220132

ABSTRACT

PURPOSE: Vascular endothelial growth factor receptor 2-blocking antibody (DC101) has inhibitory effects on tumor growth and angiogenesis in vivo. The human sodium/iodide symporter (hNIS) gene has been shown to be a useful molecular imaging reporter gene. Here, we investigated the evaluation of therapeutic efficacy by molecular imaging in reporter gene transfected tumor xenografts using a gamma imaging system. METHODS: The hNIS gene was transfected into MDA-MB-231 cells using Lipofectamine. The correlation between the number of MDA-MB-231-hNIS cells and the uptake of (99m)Tc-pertechnetate or (125)I was investigated in vitro by gamma imaging and counting. MDA-MB-231-hNIS cells were injected subcutaneously into mice. When the tumor volume reached 180-200 mm(3), we randomly assigned five animals to each of three groups representing different tumor therapies; no DC101 (control), 100 µg, or 150 µg DC101/mouse. One week and 2 weeks after the first injection of DC101, gamma imaging was performed. Mice were sacrificed 2 weeks after the first injection of DC101. The tumor tissues were used for reverse transcriptase-polymerase chain reaction (RT-PCR) and CD31 staining. RESULTS: Uptake of (125)I and (99m)Tc-pertechnetate into MDA-MB-231-hNIS cells in vitro showed correlation with the number of cells. In DC101 treatment groups, the mean tumor volume was smaller than that of the control mice. Furthermore, tumor uptake of (125)I was lower than in the controls. The CD31 staining and RT-PCR assay results showed that vessel formation and expression of the hNIS gene were significantly reduced in the tumor tissues of treatment groups. CONCLUSION: This study demonstrated the power of molecular imaging using a gamma imaging system for evaluating the therapeutic efficacy of an antitumor treatment. Molecular imaging systems may be useful in evaluation and development of effective diagnostic and/or therapeutic antibodies for specific target molecules.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , Molecular Imaging/methods , Symporters/metabolism , Vascular Endothelial Growth Factor Receptor-2/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Rats , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Tumor Burden/drug effects
10.
Bioconjug Chem ; 22(2): 186-92, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21243999

ABSTRACT

Oleic acid-conjugated chitosan (oleyl-chitosan) is a powerful platform for encapsulating oleic acid-decorated iron oxide nanoparticles (ION), resulting in a good magnetic resonance imaging (MRI) probe. Oleyl-chitosan could self-assemble into core-shell structures in aqueous solution and provide the effective core compartment for loading ION. ION-loaded oleyl-chitosan nanoparticles showed good enhanced MRI sensitivity in a MR scanner. Cy5.5 dye was accessed to the oleyl-chitosan conjugate for near-infrared (NIR) in vivo optical imaging. After intravenous injection of ION-loaded Cy5.5-conjugated oleyl-chitosan (ION-Cy5.5-oleyl-chitosan) nanoparticles in tumor-bearing mice, both NIRF and MR imaging showed the detectable signal intensity and enhancement in tumor tissues via enhanced permeability and retention (EPR) effect. Tumor accumulation of the nanoparticles was confirmed through ex vivo fluorescence images and Prussian blue staining images in tumor tissues. It is concluded that ION-Cy5.5-oleyl-chitosan nanoparticle is highly an effective imaging probe for detecting tumor in vivo.


Subject(s)
Carbocyanines/chemistry , Chitosan/chemistry , Ferric Compounds/chemistry , Magnetic Resonance Imaging , Nanoparticles/chemistry , Neoplasms, Experimental/diagnosis , Oleic Acid/chemistry , Animals , Female , Ferrocyanides/chemistry , Mice , Mice, Nude , Permeability , Staining and Labeling , Xenograft Model Antitumor Assays
11.
Bioconjug Chem ; 21(12): 2282-8, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21049983

ABSTRACT

Syntheses and evaluation of fluoroalkylated ciprofloxacin analogues are described. Among these analogues, N4'-3-fluoropropylciprofloxacin (16) showed the most efficient antibacterial activity against E. coli strains (DH5α and TOP10) and a high binding affinity for DNA gyrase of bacteria. To develop bacteria-specific infection imaging agents for positron emission tomography (PET), no-carrier-added N4-3-[¹8F]fluoropropylciprofloxacin ([¹8F]16) was prepared in two steps from N4-3-methanesufonyloxypropylciprofloxacin, resulting in a 40% radiochemical yield (decay corrected for 100 min) via the tert-alcohol media radiofluorination protocol with high radiochemical purity (> 99%) as well as high specific activity (149 ± 75 GBq/µmol). The agent was stable (> 90%), as shown by an in vitro human serum stability assay. A bacterial uptake and blocking study of [¹8F]16 using authentic compound 16 in TOP10 cells demonstrated its high specific bacterial uptake. The results suggest that this radiotracer holds promise as a useful bacterial infection radiopharmaceutical for PET imaging.


Subject(s)
Ciprofloxacin , Diagnostic Imaging/methods , Fluorine Radioisotopes , Positron-Emission Tomography/methods , Radiopharmaceuticals , Alkylation , Animals , Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Binding, Competitive , Biological Transport , Ciprofloxacin/analogs & derivatives , Ciprofloxacin/blood , Ciprofloxacin/chemical synthesis , Ciprofloxacin/pharmacology , DNA Gyrase/metabolism , Drug Stability , Escherichia coli/drug effects , Escherichia coli/growth & development , Fluorine Radioisotopes/blood , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacology , Halogenation , Humans , Isotope Labeling , Microbial Sensitivity Tests , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Rats , Rats, Sprague-Dawley , Topoisomerase II Inhibitors
12.
Nanotechnology ; 21(28): 285102, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20562480

ABSTRACT

The aim of this study was to investigate the effect of gluconic acid (GA) conjugation on the biodistribution of cysteamine-capped quantum dots (amino-QDots) in vivo. Cadmium selenide/zinc sulfide (CdSe/ZnS) was capped with cysteamine through a thiol exchange method, and different amounts of GA were conjugated to the amine groups of cysteamine via the formation of an amide bond. The emission maxima of the synthesized QDots, the amino-QDots and the GA-conjugated amine-QDots (GA-QDots) were located at 720, 600 and 610 nm, respectively. In the cell viability studies, the GA-QDots showed very low toxicity against CHO cells as compared to the cytotoxicity of the amino-QDots. The QDots were next intravenously injected into normal mice and then we performed ex vivo optical imaging. The majority of the amino-QDots were accumulated in the lung. In contrast, the GA-QDots were cleared out of the body through the kidney. Therefore, we expect that the conjugation of GA onto the amino-QDots can create opportunities for using amino-QDots for in vivo imaging.


Subject(s)
Imaging, Three-Dimensional/methods , Nanotechnology/methods , Quantum Dots , Animals , CHO Cells , Cadmium Compounds/chemistry , Cell Death , Cricetinae , Cricetulus , Female , Gluconates/chemistry , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Photoelectron Spectroscopy , Selenium Compounds/chemistry , Spectroscopy, Fourier Transform Infrared , Sulfides/chemistry , Surface Properties , Tissue Distribution , Zinc Compounds/chemistry
13.
Pharm Res ; 27(4): 712-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20182773

ABSTRACT

PURPOSE: In this work, the aim was to prepare and characterize a magnetofluorescent polymeric nanoparticle for prostate cancer imaging in vivo. METHODS: Glycol chitosan (GC) was chemically modified with N-acetyl histidine (NAHis) as a hydrophobic moiety, and bombesin (BBN) was conjugated to the hydrophobically modified GC for use in targeting gastric-releasing peptide receptors (GRPR) overexpressed in prostate cancer cells. NAHis-GC conjugates were labeled with the near-infrared (NIR) fluorophore Cy5.5 (C-NAHis-GC conjugate). RESULTS: BBN-conjugated C-NAHis-GC nanoparticles (BC-NAHis-GC nanoparticles) showed significantly higher binding to the PC3 cell surface than nanoparticles without BBN, and the cellular binding was clearly inhibited by BBN. The tumor-to-muscle ratios of C- and BC-NAHis-GC nanoparticles were 2.26 +/- 0.66 and 5.37 +/- 0.43, respectively. The tumor accumulation of BC-NAHis-GC nanoparticles was clearly reduced by co-injection of BBN. Further, iron oxide nanoparticles (IO) were loaded into BC-NAHis-GC nanoparticles to investigate the possibility of use as a probe for MRI. IO-BC-NAHis-GC nanoparticles were well observed in the PC3 cells, and the blocking with BBN significantly reduced the cellular binding of the nanoparticles. CONCLUSION: These results demonstrate that the BBN conjugation to NAHis-GC nanoparticles improves their tumor accumulation in PC3-bearing mice in comparison to nanoparticles without BBN, suggesting that BC-NAHis-GC nanoparticles may be useful for prostate cancer imaging.


Subject(s)
Bombesin , Chitosan/chemistry , Nanoparticles , Neurotransmitter Agents , Prostatic Neoplasms/diagnosis , Receptors, Bombesin/metabolism , Animals , Bombesin/chemistry , Bombesin/metabolism , Cell Line, Tumor , Female , Fluorescence , Magnetics , Male , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/metabolism
14.
J Biomed Mater Res A ; 92(4): 1510-7, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19425046

ABSTRACT

Expression of vascular endothelial growth factor receptors (VEGFRs) increases in ischemic muscles, and thus, VEGFR could potentially be used as marker to detect ischemia. Here, we investigated whether (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates could identify VEGFR-2 overexpressed in ischemia. To this end, chitosan was conjugated with the DC101 antibody and Cy5.5, FITC, or the HYNIC chelator for (99m)Tc-labeling. Targeting of the conjugate was evaluated in vitro and in vivo through cell-binding studies and gamma/optical imaging, respectively. A hindlimb ischemic mouse model was surgically created by femoral artery occlusion. The chitosan-DC101 conjugates exhibited VEGFR-selective cell binding properties as determined by both confocal microscopy and flow cytometry. At postoperative times of 2, 12, and 24 h, (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates were intravenously injected into the mice, and gamma/optical imaging studies were conducted at 1 or 3 h. Both the gamma and optical imaging results indicated a significantly higher uptake in ischemic muscles when compared with the contralateral nonischemic muscle. Further, semiquantitative analysis of scintigraphic imaging data revealed that the ischemic to contralateral limb ratio was 4.5 +/- 0.25 at 24 h postoperation. Western blotting analysis confirmed VEGFR expression in the ischemic muscle. In conclusion, we believe that (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates have the potential to be useful as VEGFR-2-targeted imaging agents for monitoring ischemia.


Subject(s)
Antibodies, Monoclonal , Chitosan , Ischemia/metabolism , Microvessels/metabolism , Molecular Imaging/methods , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Carbocyanines/chemistry , Carbocyanines/metabolism , Cells, Cultured , Chitosan/chemistry , Chitosan/metabolism , Female , Hindlimb/blood supply , Hindlimb/diagnostic imaging , Hindlimb/metabolism , Humans , Mice , Mice, Inbred BALB C , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Neovascularization, Physiologic , Radionuclide Imaging , Technetium/chemistry , Technetium/metabolism
15.
Bioconjug Chem ; 20(7): 1299-306, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19534520

ABSTRACT

Mesenchymal-epithelial transition factor (c-Met) is a receptor tyrosine kinase that has been shown to be overexpressed and mutated in a variety of malignancies, such as glioma. We have recently found that an (125)I-radiolabeled Gly-Gly-Gly (GGG)- or 8-aminooctanoic acid (AOC)-containing c-Met binding peptide (cMBP) specifically targets c-Met receptor in vivo and in vitro. In this report, cyanine dye 5.5 (Cy5.5)-conjugated GGG- or AOC-containing cMBPs were evaluated in human cancer cell xenografts in order to investigate the possibility of c-Met receptor targeting using an optical imaging system. The receptor binding affinity of Cy5.5-conjugated peptides was tested in 96-well plates coated with a c-Met/Fc chimeric protein. Optical imaging studies were performed in U87MG and Ramos bearing athymic mice. The binding affinities of Cy5.5-conjugated GGG- or AOC-containing cMBPs were determined to be 0.318 and 0.342 microM, respectively. Confocal images show that Cy5.5-conjugated peptides bound mainly to the cell surface and that peptide binding was clearly inhibited by free cMBP. Subcutaneous U87MG tumors were clearly visualized with each of the two fluorescent probes. Of the two, cMBP-AOC-Cy5.5 displayed higher tumor uptake and tumor-to-normal tissue ratios at 10 min to 24 h postinjection in the U87MG tumor model. For the in vivo blocking study, cMBP-AOC-Cy5.5 (4 nmol) was co-injected with cold cMBP (0.13 micromol) into the U87MG xenograft mice. Image-based tumoral uptake decreased up to approximately 35%. These results suggest that Cy5.5-conjugated cMBP could potentially be used to detect c-Met-positive cancers in vivo. However, additional modifications to this optical imaging agent are needed to further improve its efficacy.


Subject(s)
Diagnostic Imaging/methods , Fluorescent Dyes/chemistry , Neoplasms/diagnosis , Peptides/chemistry , Peptides/metabolism , Proto-Oncogene Proteins c-met/analysis , Animals , Caprylates/chemical synthesis , Caprylates/chemistry , Carbocyanines/chemical synthesis , Carbocyanines/chemistry , Cell Line, Tumor , Female , Fluorescent Dyes/chemical synthesis , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Microscopy, Confocal , Neoplasm Transplantation , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Peptides/chemical synthesis , Protein Binding , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics
16.
Nucl Med Biol ; 36(4): 371-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19423004

ABSTRACT

c-Met is a receptor tyrosine kinase involved in tumor cell growth, invasion, metastases and angiogenesis. Overexpression of c-Met is frequently observed in several tumor types. Here, we report the in vitro cell-binding properties and biodistribution and SPECT/CT imaging in glioma (U87MG) xenograft-bearing mice of (125)I-labeled c-Met-binding peptides (cMBPs) including analogs conjugated to amino acid and aliphatic carbon linkers. In vitro assays showed that the peptide without any linker and those with GGG and 8-aminooctanoic acid linkers had low cellular internalization and that IC(50) values of peptides were 1.5 microM, 65 nM and 85.3 nM, respectively. Biodistribution studies showed the GGG-containing peptide had higher tumor uptake and a higher tumor-to-blood activity concentration ratio than other receptor-binding ligands. SPECT/CT studies with a dedicated small-animal imaging system were performed in U87MG-bearing athymic mice. Although U87MG tumor xenografts could be visualized by SPECT/micro-CT using the various (125)I labeled cMBPs, image contrast and overall quality were unremarkable.


Subject(s)
Glioma/diagnostic imaging , Glioma/metabolism , Peptides/metabolism , Peptides/pharmacokinetics , Proto-Oncogene Proteins c-met/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Biological Transport , Blotting, Western , Caprylates/metabolism , Cell Line, Tumor , Glioma/surgery , Glycine/metabolism , Humans , Iodine Radioisotopes , Mice , Molecular Sequence Data , Peptides/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Transplantation, Heterologous
17.
Nucl Med Biol ; 36(4): 427-33, 2009 May.
Article in English | MEDLINE | ID: mdl-19423011

ABSTRACT

PURPOSE: The peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a member of the nuclear receptor superfamily of ligand-dependent transcription factors, and its role in adipogenesis and glucose metabolism has been well established. PPAR-gamma agonists have been shown to inhibit many cytokines and to have anti-inflammatory effects. In pathologic conditions, enhanced fluoro-2-deoxy-D-glucose (FDG) uptake is observed not only in malignant tumors but also in inflammatory lesions, and this uptake occurs through the glucose transporter in these cells. Thus, the present study was undertaken to investigate the potential of using PPAR-gamma's glucose uptake ability as a diagnostic tool to differentiate between macrophage and tumor cells. MATERIALS AND METHODS: Cellular uptake studies were carried out on macrophage and two tumor cell lines for comparison by using (18)F-FDG. Western blot analysis was performed to determine the expression levels of both the glucose transporter and hexokinase protein. To confirm the possibility of differentiation between tumor and inflammatory lesions using rosiglitazone based on in vitro studies, (18)F-FDG (3.7 x 10(6) Bq) uptake in A549 and RAW 264.7 xenograft mice was compared. RESULTS: The cellular uptake study findings were quite different for macrophages and tumor cells. (18)F-FDG uptakes by macrophages decreased by about 60% but was increased twofold in tumor cells after rosiglitazone treatment. Moreover, the expressions of proteins related to glucose uptake correlated well with cellular glucose accumulation in both cell types. Higher tumor uptake was observed after the injection of rosiglitazone in A549 xenograft mice (1.58+/-0.55 to 4.66+/-1.16), but no significant change of (18)F-FDG uptake was shown in RAW 264.7 xenograft mice (4.04+/-1.16 to 4.00+/-0.14). CONCLUSION: The present study demonstrates the roles of PPAR-gamma agonist on FDG uptake in macrophages and tumor cells in vitro and in vivo. Our findings suggest that rosiglitazone has the potential to increase the contrast between tumor and inflammatory lesions.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Macrophages/drug effects , Macrophages/metabolism , Neoplasms/metabolism , Neoplasms/pathology , PPAR gamma/agonists , Animals , Biological Transport/drug effects , Cell Line, Tumor , Dimethyl Sulfoxide/pharmacology , Gene Expression Regulation , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Hexokinase/metabolism , Humans , Inflammation/pathology , Mice , Neoplasms/diagnostic imaging , Positron-Emission Tomography , Rosiglitazone , Thiazolidinediones/pharmacology , Tomography, X-Ray Computed
18.
Int J Pharm ; 372(1-2): 169-76, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19429277

ABSTRACT

The goal of this study was to develop a gene delivery imaging system that targets hepatocytes to help diagnose and treat various liver diseases. To this end, we prepared superparamagnetic iron oxide nanoparticles (SPIO)-loaded with water-soluble chitosan (WSC)-linoleic acid (LA) nanoparticles (SCLNs) that formed gene complexes capable of localizing specifically to hepatocytes. We confirmed that (99m)Tc-labeled SCLNs delivered into mice via intravenous injection accumulated mainly in the liver using nuclear and magnetic resonance imaging. SCLN/enhanced green fluorescence protein (pEGFP) complexes were also successfully formed and were characterized with a gel retardation assay. SCLN/pEGFP complexes were transfected into primary hepatocytes, where GFP expression was observed in the cytoplasm. In addition, the injection of the gene complexes into mice resulted in significantly increased expression of GFP in hepatocytes in vivo. Furthermore, gene silencing was effectively achieved by administration of gene complexes loaded with specific siRNAs. In conclusion, our results indicate that the SCLNs have the potential to be useful for hepatocyte-targeted imaging and effective gene delivery into hepatocytes.


Subject(s)
Chitosan/administration & dosage , Ferric Compounds/administration & dosage , Gene Transfer Techniques , Hepatocytes/metabolism , Linoleic Acid/administration & dosage , Nanoparticles/administration & dosage , Animals , Cells, Cultured , Female , Gene Targeting/methods , Hepatocytes/drug effects , Hepatocytes/physiology , Magnetics , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...