Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Appl Radiat Isot ; 108: 1-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26656427

ABSTRACT

INTRODUCTION: Apoptosis has been suggested as a cytocidal mechanism of the HSV1-tk-expressing cells when exposed to ganciclovir (GCV). This study evaluated the efficacy of (111)In-labeled Annexin V for monitoring tumor responses during prodrug activation gene therapy with HSV1-tk and GCV. MATERIALS AND METHODS: Annexin V was conjugated to DOTA using N-hydroxysulfosuccinimide (sulfo-NHS) and 1-ethyl-3-[3-(dimethylamino)propyl]carbodiimide (EDC), labeled with (111)In-InCl3 and purified using size exclusion chromatography to give (111)In-DOTA-Annexin V conjugate. The radiochemical yield and the radiochemical purity of (111)In-DOTA-Annexin V were 74±12% and 98±3%, respectively (n=10). (111)In-DOTA-BSA was prepared similarly. An in vitro study to demonstrate the apoptosis of NG4TL4-STK cells after GCV treatment has been performed. Mice bearing NG4TL4-STK and NG4TL4-WT tumors were treated with GCV (10 mg/kg daily) by i.p. injection for 7 consecutive days. Before and during the GCV treatment, biodistribution studies and scintigraphic imaging were performed at 2h post injection of the radiotracers. RESULTS: The uptake of (111)In-DOTA-Annexin V in treated cells (13.41±1.30%) was 4.1 times higher than that in untreated cells (3.21±0.37%). The GCV-induced cell apoptosis in NG4TL4-STK tumor resulted in a significantly increasing accumulation of (111)In-DOTA-Annexin V (1.92±0.32%ID/g at day 0, 4.79±0.86%ID/g at day 2, 4.56±0.58%ID/g at day 4) was observed, but not for that of (111)In-DOTA-BSA. During consecutive GCV treatment, scintigraphic imaging with (111)In-DOTA-Annexin V revealed high uptake in NG4TL4-STK tumor compared with that in NG4TL4-WT tumor. However, no specific (111)In-DOTA-BSA accumulation in NG4TL4-STK and NG4TL4-WT tumors was observed throughout the course of GCV treatment. CONCLUSIONS: This study demonstrated that (111)In-DOTA-Annexin V can be used for monitoring tumor cell apoptosis during prodrug activation gene therapy with HSV1-tk and GCV for cancer treatment.


Subject(s)
Annexin A5/pharmacokinetics , Coordination Complexes/pharmacokinetics , Genetic Therapy/methods , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Prodrugs/administration & dosage , Sarcoma/diagnostic imaging , Sarcoma/therapy , Animals , Annexin A5/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/chemistry , Drug Monitoring/methods , Ganciclovir/administration & dosage , Herpesvirus 1, Human/enzymology , Heterocyclic Compounds, 1-Ring/chemistry , Isotope Labeling/methods , Metabolic Clearance Rate , Mice , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Sarcoma/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tissue Distribution , Treatment Outcome
2.
Biomed Res Int ; 2014: 912498, 2014.
Article in English | MEDLINE | ID: mdl-25254219

ABSTRACT

INTRODUCTION: Benzamide can specifically bind to melanoma cells. A 18F-labeled benzamide derivative, [18F]N-(2-diethylaminoethyl)-4-[2-(2-(2-fluoroethoxy) ethoxy)ethoxy]benzamide ([18F]FPBZA), was developed as a promising PET probe for primary and metastatic melanoma. METHODS: [18F]FPBZA was synthesized via a one-step radiofluorination in this study. The specific uptake of [18F]FPBZA was studied in B16F0 melanoma cells, A375 amelanotic melanoma cells, and NB-DNJ-pretreated B16F0 melanoma cells. The biological characterization of [18F]FPBZA was performed on mice bearing B16F0 melanoma, A375 amelanotic melanoma, or inflammation lesion. RESULTS: [18F]FPBZA can be prepared efficiently with a yield of 40-50%. The uptake of [18F]FPBZA by B16F0 melanoma cells was significantly higher than those by A375 tumor cells and NB-DNJ-pretreated B16F0 melanoma cells. B16F0 melanoma displayed prominent uptake of [18F]FPBZA at 2 h (7.81±0.82%ID/g), compared with A375 tumor and inflammation lesion (3.00±0.71 and 1.67±0.56%ID/g, resp.). [18F]FPBZA microPET scan clearly delineated B16F0 melanoma but not A375 tumor and inflammation lesion. In mice bearing pulmonary metastases, the lung radioactivity reached 4.77±0.36%ID/g at 2 h (versus 1.16±0.23%ID/g in normal mice). CONCLUSIONS: Our results suggested that [18F]FPBZA PET would provide a promising and specific approach for the detection of primary and metastatic melanoma lesions.


Subject(s)
Benzamides , Melanoma, Experimental/diagnosis , Positron-Emission Tomography/methods , Radiopharmaceuticals , Animals , Benzamides/chemical synthesis , Benzamides/chemistry , Fluorodeoxyglucose F18/chemical synthesis , Fluorodeoxyglucose F18/chemistry , Humans , Melanoma, Experimental/pathology , Mice , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Tissue Distribution
3.
Ultrasonics ; 53(2): 320-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22832082

ABSTRACT

OBJECTIVE: In this study, albumin-shelled, targeted MBs (tMBs) were first demonstrated with the expectation of visualization of biodistribution of albumin-shelled tMBs. The actual biodistribution of albumin-shelled tMBs is of vital importance either for molecular imaging or for drug delivery. MOTIVATION: Recently, albumin microbubbles (MBs) have been studied for drug and gene delivery in vitro and in vivo through cavitation. Targeted lipid-shelled MBs have been applied for ultrasound molecular imaging and conjugated with radiolabeled antibodies for whole-body biodistribution evaluations. The novelty of the work is that, in addition to the lipid tMBs, the albumin tMBs was also applied in biodistribution detection. METHODS: Multimodality albumin-shelled, (18)F-SFB-labeled VEGFR2 tMBs were synthesized, and their characteristics in mice bearing MDA-MB-231 human breast cancer were investigated with micro-positron-emission tomography (microPET) and high-frequency ultrasound (microUS). RESULTS: Albumin-shelled MBs can be labeled with (18)F-SFB directly and conjugated with antibodies for dual molecular imaging. The albumin-shelled tMBs show a lifetime in 30min in the blood pool and a highly specific adherence to tumor vessels in mice bearing human breast cancer. CONCLUSIONS: From the evaluations of whole-body biodistribution, the potential of the dual molecular imaging probe for drug or gene delivery in animal experiments with albumin shelled MBs has been investigated.


Subject(s)
Albumins , Contrast Media , Fluorine Radioisotopes , Fluorocarbons , Mammary Neoplasms, Experimental/diagnostic imaging , Microbubbles , Positron-Emission Tomography , Ultrasonography , Animals , Female , Isotope Labeling , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/analysis
4.
Nucl Med Biol ; 39(7): 1026-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22762865

ABSTRACT

INTRODUCTION: (99m)Tc-labeled macroaggregated albumin ((99m)Tc-MAA) scintigraphy scan is routinely performed for lung perfusion imaging and for the assessment of in vivo distribution of (90)Y-labeled SIR-Spheres prior to selective internal radiation treatment for hepatocellular carcinoma. Positron emission tomography (PET) imaging is superior to gamma scintigraphy in terms of sensitivity, spatial resolution and accuracy of quantification. This study reported that (18)F-labeled macroaggregated albumin ((18)F-MAA) is an ideal PET imaging surrogate for (99m)Tc-MAA. METHODS: (18)F-MAA was prepared from the commercial MAA kit via a one-step conjugation with N-succinimidyl 4-(18)F-fluorobenzoate ((18)F-SFB). The biodistribution study and microPET/microSPECT imaging were conducted in normal SD rats after intravenous injection of (18)F-MAA/(99m)Tc-MAA. A comparison study of these two radiotracers was performed after co-injection via the intrahepatic arterial in a N1S1 hepatoma-bearing SD rat model. RESULTS: The optimal condition for (18)F-MAA preparation is coupling MAA (0.5mg) with (18)F-SFB at 45°C for 5 min in a phosphate buffer of pH 8.5. (18)F-MAA was prepared in 60 min with high radiochemical yield (30%-35%) and high radiochemical purity (>95%). The in vivo distribution of (18)F-MAA after intravenous injection meets the specifications of MAA depicted in European Pharmacopeia. Our study demonstrated excellent correlation between (18)F-MAA and (99m)Tc-MAA in the regional distribution of tumor, liver and lungs (R(2)=0.965, 0.886 and 0.991, respectively), and also in the tumor-to-liver and tumor-to-lungs ratio (R(2)=0.965 and 0.987, respectively) in a N1S1 hepatoma-bearing SD rat model. The organ uptakes derived from animal PET/CT and SPECT/CT imaging after administration of these two tracers were in accordance with those obtained in the distribution studies. CONCLUSIONS: Starting from commercial MAA kit, an efficient preparation of (18)F-MAA was successfully established. Highly correlated, almost parallel, regional distribution of (18)F-MAA and (99m)Tc-MAA in both normal rats and hepatoma-bearing rats was observed. The findings, taken together, demonstrate that (18)F-MAA is an ideal surrogate for (99m)Tc-MAA for clinical PET applications.


Subject(s)
Fluorine Radioisotopes , Positron-Emission Tomography/methods , Sulfhydryl Compounds , Technetium Tc 99m Aggregated Albumin , Animals , Benzoates/chemistry , Carcinoma, Hepatocellular/diagnostic imaging , Cell Line, Tumor , Male , Radiochemistry , Rats , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacokinetics , Technetium Tc 99m Aggregated Albumin/chemistry , Technetium Tc 99m Aggregated Albumin/pharmacokinetics , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
5.
Phys Med Biol ; 57(9): 2787-802, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22510713

ABSTRACT

This paper presents new albumin-shelled Gd-DTPA microbubbles (MBs) that can concurrently serve as a dual-modality contrast agent for ultrasound (US) imaging and magnetic resonance (MR) imaging to assist blood-brain barrier (BBB) opening and detect intracerebral hemorrhage (ICH) during focused ultrasound brain drug delivery. Perfluorocarbon-filled albumin-(Gd-DTPA) MBs were prepared with a mean diameter of 2320 nm and concentration of 2.903×10(9) MBs ml(-1) using albumin-(Gd-DTPA) and by sonication with perfluorocarbon (C(3)F(8)) gas. The albumin-(Gd-DTPA) MBs were then centrifuged and the procedure was repeated until the free Gd(3+) ions were eliminated (which were detected by the xylenol orange sodium salt solution). The albumin-(Gd-DTPA) MBs were also characterized and evaluated both in vitro and in vivo by US and MR imaging. Focused US was used with the albumin-(Gd-DTPA) MBs to induce disruption of the BBB in 18 rats. BBB disruption was confirmed with contrast-enhanced T(1)-weighted turbo-spin-echo sequence MR imaging. Heavy T(2)*-weighted 3D fast low-angle shot sequence MR imaging was used to detect ICH. In vitro US imaging experiments showed that albumin-(Gd-DTPA) MBs can significantly enhance the US contrast in T(1)-, T(2)- and T(2)*-weighted MR images. The r(1) and r(2) relaxivities for Gd-DTPA were 7.69 and 21.35 s(-1)mM(-1), respectively, indicating that the MBs represent a positive contrast agent in T(1)-weighted images. In vivo MR imaging experiments on 18 rats showed that focused US combined with albumin-(Gd-DTPA) MBs can be used to both induce disruption of the BBB and detect ICH. To compare the signal intensity change between pure BBB opening and BBB opening accompanying ICH, albumin-(Gd-DTPA) MB imaging can provide a ratio of 5.14 with significant difference (p = 0.026), whereas Gd-DTPA imaging only provides a ratio of 2.13 and without significant difference (p = 0.108). The results indicate that albumin-(Gd-DTPA) MBs have potential as a US/MR dual-modality contrast agent for BBB opening and differentiating focused-US-induced BBB opening from ICH, and can monitor the focused ultrasound brain drug delivery process.


Subject(s)
Albumins/chemistry , Blood-Brain Barrier/metabolism , Fluorocarbons/chemistry , Gadolinium DTPA , Magnetic Resonance Imaging/methods , Microspheres , Ultrasonography/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Contrast Media , Drug Delivery Systems , Feasibility Studies , Gadolinium DTPA/chemistry , Humans , Magnetic Phenomena , Male , Permeability , Rats , Rats, Sprague-Dawley
6.
Clin Cancer Res ; 18(1): 238-47, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22019516

ABSTRACT

PURPOSE: To develop an imaging technology for protease activities in patients that could help in prognosis prediction and in design of personalized, protease-based inhibitors and prodrugs for targeted therapy. EXPERIMENTAL DESIGN: Polyethylene glycol (PEG) was covalently attached to the N-terminus of a hydrophilic peptide substrate (GPLGVR) for matrix metalloproteinase (MMP) to increase hydrophilicity. PEG-peptide was then linked to a hydrophobic tetramethylrhodamine (TMR) domain and labeled with (18)F to form a PEG-peptide-(18)F-TMR probe. Specific cleavage of the probe by MMP2 was tested in vitro by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF). In vivo imaging of MMP2-expressing tumors was evaluated by micro-PET. RESULTS: The hydrophobic TMR fragment (948 Da) was specifically generated by MMP2 enzymes and MMP-expressing HT1080 cells but not control MCF-7 cells. MMP-expressing HT1080 cells and tumors selectively accumulated the hydrolyzed, hydrophobic TMR fragment at sites of protease activity. Importantly, we found that (18)F-labeled probe ((18)F-TMR) preferentially localized in HT1080 tumors but not control MCF-7 tumors as shown by micro-PET. Uptake of the probe in HT1080 tumors was 18.4 ± 1.9-fold greater than in the MCF-7 tumors 30 minutes after injection. These results suggest that the PEG-peptide-(18)F-TMR probe displays high selectivity for imaging MMP activity. CONCLUSIONS: This strategy successfully images MMP expression in vivo and may be extended to other proteases to predict patient prognosis and to design personalized, protease-based inhibitors and prodrug-targeted therapies.


Subject(s)
Adenocarcinoma/diagnostic imaging , Breast Neoplasms/diagnostic imaging , Diagnostic Imaging , Fibrosarcoma/diagnostic imaging , Matrix Metalloproteinase Inhibitors , Peptide Fragments/metabolism , Positron-Emission Tomography , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Fragments/pharmacokinetics , Polyethylene Glycols/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution
7.
Article in English | MEDLINE | ID: mdl-21622052

ABSTRACT

The use of pulsed high-intensity focused ultrasound (HIFU) with an ultrasound contrast agent (UCA) has been shown to disrupt the blood-brain barrier (BBB) noninvasively and reversibly in the targeted regions. This study evaluated the relative permeability of the blood-tumor barrier (BTB) after sonication by pulsed HIFU. Entry into the brain of chemotherapeutic agents is impeded by the BBB even though the permeability of this barrier may be partially reduced in the presence of a brain tumor. F98 glioma-bearing rats were injected intravenously with Evans blue (EB) with or without BTB disruption induced by pulsed HIFU. Sonication was applied at an ultrasound frequency of 1 MHz with a 5% duty cycle, and a repetition frequency of 1 Hz. The accumulation of EB in brain tumor and the tumor-to-contralateral brain ratio of EB were highest after pulsed HIFU exposure. Sonication followed by EB injection showed a tumor-to-contralateral brain ratio in the target tumors which was about 2 times that of the control tumors. This research demonstrates that pulsed HIFU enhances the relative permeability of the BTB in glioma- bearing rats. The results of this pilot study support the idea that further evaluation of other treatment strategies, such as HIFU exposure in addition to combined chemotherapy or repeated pulsed HIFU exposure to increase delivery of drugs into brain tumors, might be useful.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/radiation effects , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Glioma/diagnostic imaging , Glioma/therapy , High-Intensity Focused Ultrasound Ablation/methods , Animals , Body Weight , Brain Chemistry/radiation effects , Brain Neoplasms/blood supply , Capillary Permeability/radiation effects , Disease Models, Animal , Drug Delivery Systems , Evans Blue , Glioma/blood supply , Histocytochemistry , Magnetic Resonance Imaging , Male , Rats , Rats, Inbred F344 , Ultrasonography
8.
Bioconjug Chem ; 22(5): 938-48, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21443266

ABSTRACT

Antibody-directed enzyme prodrug therapy (ADEPT) utilizing ß-glucuronidase is a promising method to enhance the therapeutic index of cancer chemotherapy. In this approach, an immunoenzyme (antibody-ß-glucuronidase fusion protein) is employed to selectively activate anticancer glucuronide prodrugs in the tumor microenvironment. A major roadblock to the clinical translation of this therapeutic strategy, however, is the low enzymatic activity and strong immunogenicity of the current generation of immunoenzymes. To overcome this problem, we fused a humanized single-chain antibody (scFv) of mAb CC49 to S2, a human ß-glucuronidase (hßG) variant that displays enhanced catalytic activity for prodrug hydrolysis. Here, we show that hcc49-S2 displayed 100-fold greater binding avidity than hcc49 scFv, possessed greater enzymatic activity than wild-type hßG, and more effectively killed antigen-positive cancer cells exposed to an anticancer glucuronide prodrug as compared to an analogous hßG immunoenzyme. Treatment of tumor-bearing mice with hcc49-S2 followed by prodrug significantly delayed tumor growth as compared to hcc49-hßG. Our study shows that hcc49-S2 is a promising targeted enzyme for cancer treatment and demonstrates that enhancement of human enzyme catalytic activity is a powerful approach to improve immunoenzyme efficacy.


Subject(s)
Antibodies, Neoplasm/metabolism , Glucuronidase/metabolism , Glucuronides/metabolism , Prodrugs/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Microenvironment , Animals , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/isolation & purification , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biocatalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glucuronidase/chemistry , Glucuronidase/isolation & purification , Glucuronides/pharmacology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Molecular Imaging , NIH 3T3 Cells , Prodrugs/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Nucl Med Biol ; 32(4): 367-75, 2005 May.
Article in English | MEDLINE | ID: mdl-15878506

ABSTRACT

UNLABELLED: 2-Deoxy-2-[(18)F]fluoro-d-glucose ([(18)F]FDG) has been extensively used as positron emission tomography (PET) tracer in clinical tumor imaging. This study compared the pharmacokinetics of two (18)F-labeled amino acid derivatives, O-2-[(18)F]fluoroethyl-l-tyrosine (l-[(18)F]FET) and 4-borono-2-[(18)F]fluoro-l-phenylalanine-fructose (l-[(18)F]FBPA-Fr), to that of [(18)F]FDG in an animal brain tumor model. METHODS: A self-modified automated PET tracer synthesizer was used to produce no-carrier-added (nca) l-[(18)F]FET. The cellular uptake, biodistribution, autoradiography and microPET imaging of l-[(18)F]FET, l-[(18)F]FBPA-Fr and [(18)F]FDG were performed with F98 glioma cell culture and F98 glioma-bearing Fischer344 rats. RESULTS: The radiochemical purity of l-[(18)F]FET was >98% and the radiochemical yield was 50% in average of 16 runs. The uptake of l-[(18)F]FET and l-[(18)F]FBPA-Fr in the F98 glioma cells increased rapidly for the first 5 min and reached a steady-state level after 10 min of incubation, whereas the cellular uptake of [(18)F]FDG kept increasing during the study period. The biodistribution of l-[(18)F]FET, l-[(18)F]FBPA-Fr and [(18)F]FDG in the brain tumors was 1.26+/-0.22, 0.86+/-0.08 and 2.77+/-0.44 %ID/g at 60 min postinjection, respectively, while the tumor-to-normal brain ratios of l-[(18)F]FET (3.15) and l-[(18)F]FBPA-Fr (3.44) were higher than that of [(18)F]FDG (1.44). Both microPET images and autoradiograms of l-[(18)F]FET and l-[(18)F]FBPA-Fr exhibited remarkable uptake with high contrast in the brain tumor, whereas [(18)F]FDG showed high uptake in the normal brain and gave blurred brain tumor images. CONCLUSION: Both l-[(18)F]FET and l-[(18)F]FBPA-Fr are superior to [(18)F]FDG for the brain tumor imaging as shown in this study with microPET.


Subject(s)
Boron Compounds/pharmacokinetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Glioma/diagnostic imaging , Glioma/metabolism , Glycopeptides/pharmacokinetics , Positron-Emission Tomography/methods , Tyrosine/analogs & derivatives , Animals , Brain/diagnostic imaging , Brain/metabolism , Cell Line, Tumor , Disease Models, Animal , Fluorine Radioisotopes/pharmacokinetics , Metabolic Clearance Rate , Molecular Probe Techniques , Organ Specificity , Radiopharmaceuticals/pharmacokinetics , Rats , Tissue Distribution , Tyrosine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...