Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Eur J Surg Oncol ; 47(3 Pt B): 628-634, 2021 03.
Article in English | MEDLINE | ID: mdl-33069505

ABSTRACT

BACKGROUND: Approximately 15% of patients undergoing resection for presumed perihilar cholangiocarcinoma (PHC) have benign disease at final pathological assessment. Molecular imaging targeting tumor-specific biomarkers could serve as a novel diagnostic tool to reduce these futile surgeries. Imaging agents have been developed, selectively binding integrin ανß6, a cell receptor upregulated in pancreatobiliary malignancies, for both (preoperative) PET and (intraoperative) fluorescent imaging. Here, expression of integrin ανß6 is evaluated in PHC, intrahepatic cholangiocarcinoma (ICC), hepatocellular carcinoma (HCC) and benign disease mimicking PHC using immunohistochemistry. MATERIALS & METHODS: Three tissue microarrays (TMA) including 103 PHC tumor cores and sixty tissue samples were selected from resection specimens of pathologically proven PHC (n = 20), ICC (n = 10), HCC (n = 10), metastatic PHC lymph nodes (n = 10) and benign disease (presumed PHC with benign disease at pathological assessment, n = 10). These samples were stained for integrin ανß6 and quantified using the H-score. RESULTS: Immunohistochemical staining for integrin ανß6 showed membranous expression in all twenty PHC whole mount slides (100%) and 93 out of 103 (92%) PHC tumor cores. Mean H-score of PHC samples was 195 ± 71, compared to a mean H-score of 126 ± 57 in benign samples (p = 0.013). In both benign and PHC samples, inflammatory infiltrates and pre-existent peribiliary glands showed integrin ανß6 expression. The mean H-score across ten ICC was 33 ± 53, which was significantly lower compared to PHC (p < 0.001) but too weak to consistently discriminate ICC from HCC (H-score 0)(p = 0.062). CONCLUSION: Integrin ανß6 is abundantly expressed in PHC and associated metastatic lymph nodes. Expression is significantly higher in PHC as compared to benign disease mimicking PHC, ICC and HCC, emphasizing its potential as a target for tumor-specific molecular imaging.


Subject(s)
Antigens, Neoplasm/metabolism , Bile Duct Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , Cholangiocarcinoma/metabolism , Integrins/metabolism , Klatskin Tumor/metabolism , Liver Neoplasms/metabolism , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/pathology , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Klatskin Tumor/diagnosis , Klatskin Tumor/pathology , Liver Diseases/diagnosis , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Male , Middle Aged , Molecular Imaging , Optical Imaging , Positron Emission Tomography Computed Tomography , Tissue Array Analysis
2.
Photoacoustics ; 19: 100196, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32612928
3.
Cancer Res ; 74(24): 7475-86, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25320008

ABSTRACT

Histone deacetylases (HDAC) that regulate gene expression are being explored as cancer therapeutic targets. In this study, we focused on HDAC6 based on its ability to inhibit cancerous Hsp90 chaperone activities by disrupting Hsp90/p23 interactions. To identify novel HDAC6 inhibitors, we used a dual-luciferase reporter system in cell culture and living mice by bioluminescence imaging (BLI). On the basis of existing knowledge, a library of hydrazone compounds was generated for screening by coupling cinnamic hydroxamates with aldehydes and ketones. Potency and selectivity were determined by in vitro HDAC profiling assays, with further evaluation to inhibit Hsp90(α/ß)/p23 interactions by BLI. In this manner, we identified compound 1A12 as a dose-dependent inhibitor of Hsp90(α/ß)/p23 interactions, UKE-1 myeloid cell proliferation, p21(waf1) upregulation, and acetylated histone H3 levels. 1A12 was efficacious in tumor xenografts expressing Hsp90(α)/p23 reporters relative to carrier control-treated mice as determined by BLI. Small animal (18)F-FDG PET/CT imaging on the same cohort showed that 1A12 also inhibited glucose metabolism relative to control subjects. Ex vivo analyses of tumor lysates showed that 1A12 administration upregulated acetylated-H3 by approximately 3.5-fold. Taken together, our results describe the discovery and initial preclinical validation of a novel selective HDAC inhibitor.


Subject(s)
Histone Deacetylase Inhibitors/isolation & purification , Hydroxamic Acids/isolation & purification , Molecular Imaging , Multimodal Imaging , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cinnamates/chemical synthesis , Cinnamates/isolation & purification , Cinnamates/pharmacology , Fluorodeoxyglucose F18 , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Humans , Hydroxamic Acids/chemical synthesis , Mice , Myeloid Cells/drug effects
4.
Cancer Res ; 74(4): 979-1004, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24514041

ABSTRACT

Photoacoustic imaging (PAI) has the potential for real-time molecular imaging at high resolution and deep inside the tissue, using nonionizing radiation and not necessarily depending on exogenous imaging agents, making this technique very promising for a range of clinical applications. The fact that PAI systems can be made portable and compatible with existing imaging technologies favors clinical translation even more. The breadth of clinical applications in which photoacoustics could play a valuable role include: noninvasive imaging of the breast, sentinel lymph nodes, skin, thyroid, eye, prostate (transrectal), and ovaries (transvaginal); minimally invasive endoscopic imaging of gastrointestinal tract, bladder, and circulating tumor cells (in vivo flow cytometry); and intraoperative imaging for assessment of tumor margins and (lymph node) metastases. In this review, we describe the basics of PAI and its recent advances in biomedical research, followed by a discussion of strategies for clinical translation of the technique.


Subject(s)
Molecular Imaging/methods , Neoplasms/diagnosis , Photoacoustic Techniques , Translational Research, Biomedical/trends , Animals , Coloring Agents , Contrast Media , Diagnostic Techniques, Obstetrical and Gynecological , Diagnostic Techniques, Urological , Early Detection of Cancer/methods , Female , Humans , Male
5.
Gene Ther ; 20(10): 1006-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23719066

ABSTRACT

In hepatocellular carcinoma (HCC), tumor specificity of gene therapy is of utmost importance to preserve liver function. MicroRNAs (miRNAs) are powerful negative regulators of gene expression and many are downregulated in human HCC. We identified seven miRNAs that are also downregulated in tumors in a rat hepatoma model (P<0.05) and attempted to improve tumor specificity by constructing a panel of luciferase-expressing vectors containing binding sites for these miRNAs. Attenuation of luciferase expression by the corresponding miRNAs was confirmed across various cell lines and in mouse liver. We then tested our vectors in tumor-bearing rats and identified two miRNAs, miR-26a and miR-122, that significantly decreased expression in liver compared with the control vector (6.40 and 0.26%, respectively; P<0.05). In tumor, miR-122 had a nonsignificant trend towards decreased (∼50%) expression, whereas miR-26 had no significant effect on tumor expression. To our knowledge, this is the first work using differentially expressed miRNAs to de-target transgene expression in an orthotopic hepatoma model and to identify miR-26a, in addition to miR-122, for de-targeting liver. Considering the heterogeneity of miRNA expression in human HCC, this information will be important in guiding development of more personalized vectors for the treatment of this devastating disease.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genetic Vectors , Liver Neoplasms/genetics , MicroRNAs/genetics , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line , Female , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , HEK293 Cells , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Liver Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Organ Specificity , Rats , Rats, Inbred BUF , Transgenes
6.
Nano Lett ; 11(10): 4029-36, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21846107

ABSTRACT

Gold has been used as a therapeutic agent to treat a wide variety of rheumatic diseases including psoriatic arthritis, juvenile arthritis, and discoid lupus erythematosus. Although the use of gold has been largely superseded by newer drugs, gold nanoparticles are being used effectively in laboratory based clinical diagnostic methods while concurrently showing great promise in vivo either as a diagnostic imaging agent or a therapeutic agent. For these reasons, gold nanoparticles are therefore well placed to enter mainstream clinical practice in the near future. Hence, the present review summarizes the chemistry, pharmacokinetics, biodistribution, metabolism, and toxicity of bulk gold in humans based on decades of clinical observation and experiments in which gold was used to treat patients with rheumatoid arthritis. The beneficial attributes of gold nanoparticles, such as their ease of synthesis, functionalization, and shape control are also highlighted demonstrating why gold nanoparticles are an attractive target for further development and optimization. The importance of controlling the size and shape of gold nanoparticles to minimize any potential toxic side effects is also discussed.


Subject(s)
Autoimmune Diseases/drug therapy , Gold/therapeutic use , Metal Nanoparticles , Biological Availability , Gold/adverse effects , Gold/chemistry , Gold/pharmacokinetics , Humans , Tissue Distribution
7.
Gene Ther ; 18(6): 606-12, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21307888

ABSTRACT

Ideal cancer gene therapies should have high tumor specificity and efficacy, and allow systemic administration to target metastases. We recently developed a bi-directional, two-step transcriptional amplification (TSTA) system driven by the tumor-specific Survivin promoter (pSurv) to amplify the correlated expression of both the reporter gene firefly luciferase (FL) and therapeutic gene tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Here, we compare the specificity and potency of an adenovirus carrying this system (Ad-pSurv-TSTA-TRAIL-FL) to a nonspecific vector (Ad-pCMV-FL) in an orthotopic hepatocellular carcinoma (HCC) rat model after systemic administration. At 24 h after injection of Ad-pCMV-FL, bioluminescence imaging revealed a trend (P=0.30) towards greater FL expression in liver versus tumor. In striking contrast, Ad-pSurv-TSTA-TRAIL-FL showed increased FL activity within the tumor compared with the liver (P<0.01), a strong trend towards reduced liver expression compared with Ad-pCMV-FL (P=0.07), and importantly, similar FL levels within tumor compared with Ad-pCMV-FL (P=0.32). Hence, this vector shows potent, tumor-specific transgene expression even after extensive liver transduction and may be of significant value in avoiding hepatotoxicity in HCC patients. Future studies will explore the benefits of tumor-specific TRAIL expression in this model, the potential to target metastases and the extension of this vector for the treatment of other Survivin-positive tumors is warranted.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Microtubule-Associated Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Adenoviridae/genetics , Animals , Gene Expression , Gene Targeting , Genes, Reporter , Genetic Vectors , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Promoter Regions, Genetic , Rats , Sensitivity and Specificity , Survivin , Transgenes
8.
Clin Radiol ; 65(7): 500-16, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20541650

ABSTRACT

In vivo molecular imaging has a great potential to impact medicine by detecting diseases in early stages (screening), identifying extent of disease, selecting disease- and patient-specific treatment (personalized medicine), applying a directed or targeted therapy, and measuring molecular-specific effects of treatment. Current clinical molecular imaging approaches primarily use positron-emission tomography (PET) or single photon-emission computed tomography (SPECT)-based techniques. In ongoing preclinical research, novel molecular targets of different diseases are identified and, sophisticated and multifunctional contrast agents for imaging these molecular targets are developed along with new technologies and instrumentation for multi-modality molecular imaging. Contrast-enhanced molecular ultrasound (US) with molecularly-targeted contrast microbubbles is explored as a clinically translatable molecular imaging strategy for screening, diagnosing, and monitoring diseases at the molecular level. Optical imaging with fluorescent molecular probes and US imaging with molecularly-targeted microbubbles are attractive strategies as they provide real-time imaging, are relatively inexpensive, produce images with high spatial resolution, and do not involve exposure to ionizing irradiation. Raman spectroscopy/microscopy has emerged as a molecular optical imaging strategy for ultrasensitive detection of multiple biomolecules/biochemicals with both in vivo and ex vivo versatility. Photoacoustic imaging is a hybrid of optical and US techniques involving optically-excitable molecularly-targeted contrast agents and quantitative detection of resulting oscillatory contrast agent movement with US. Current preclinical findings and advances in instrumentation, such as endoscopes and microcatheters, suggest that these molecular imaging methods have numerous potential clinical applications and will be translated into clinical use in the near future.


Subject(s)
Contrast Media , Disease/genetics , Molecular Imaging/trends , Precision Medicine/trends , Humans , Molecular Imaging/methods , Neoplasms/diagnosis , Neoplasms/genetics , Precision Medicine/methods , Reproducibility of Results
9.
Gene Ther ; 17(7): 827-38, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20237511

ABSTRACT

Transcriptional targeting for cardiac gene therapy is limited by the relatively weak activity of most cardiac-specific promoters. We have developed a bidirectional plasmid vector, which uses a two-step transcriptional amplification (TSTA) strategy to enhance the expression of two optical reporter genes, firefly luciferase (fluc) and Renilla luciferase (hrluc), driven by the cardiac troponin T (cTnT) promoter. The vector was characterized in vitro and in living mice using luminometry and bioluminescence imaging to assess its ability to mediate strong, correlated reporter gene expression in a cardiac cell line and the myocardium, while minimizing expression in non-cardiac cell lines and the liver. In vitro, the TSTA system significantly enhanced cTnT-mediated reporter gene expression with moderate preservation of cardiac specificity. After intramyocardial and hydrodynamic tail vein delivery of an hrluc-enhanced variant of the vector, long-term fluc expression was observed in the heart, but not in the liver. In both the cardiac cell line and the myocardium, fluc expression correlated well with hrluc expression. These results show the vector's ability to effectively amplify and couple transgene expression in a cardiac-specific manner. Further replacement of either reporter gene with a therapeutic gene should allow non-invasive imaging of targeted gene therapy in living subjects.


Subject(s)
Gene Amplification , Gene Targeting , Gene Transfer Techniques , Genetic Vectors , Plasmids , Promoter Regions, Genetic , Transgenes , Troponin/genetics , Animals , Cell Line , Female , Genes, Reporter , Liver/metabolism , Luciferases, Firefly/genetics , Luciferases, Renilla/genetics , Mice , Mice, Inbred BALB C , Myocardium/metabolism , Transcription, Genetic
10.
Gene Ther ; 16(4): 547-57, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19092860

ABSTRACT

Human adipose tissue mesenchymal stromal cells (AMSCs) share common traits, including similar differentiation potential and cell surface markers, with their bone marrow counterparts. Owing to their general availability, higher abundance and ease of isolation AMSCs may be convenient autologous delivery vehicles for localized tumor therapy. We demonstrate a model for tumor therapy development based on the use of AMSCs expressing renilla luciferase and thymidine kinase, as cellular vehicles for ganciclovir-mediated bystander killing of firefly luciferase expressing tumors, and noninvasive bioluminescence imaging to continuously monitor both, tumor cells and AMSCs. We show that the therapy delivering AMSCs survive long time within tumors, optimize the ratio of AMSCs to tumor cells for therapy, and asses the therapeutic effect in real time. Treatment of mice bearing prostate tumors plus therapeutic AMSCs with the prodrug ganciclovir induced bystander killing effect, reducing the number of tumor cells to 1.5 % that of control tumors. Thus, AMSCs could be useful vehicles to deliver localized therapy, with potential for clinical application in inoperable tumors and surgical borders after tumor resection. This approach, useful to evaluate efficiency of therapeutic models, should facilitate the selection of cell types, dosages, therapeutic agents and treatment protocols for cell-based therapies of specific tumors.


Subject(s)
Bystander Effect , Genetic Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Prostatic Neoplasms/therapy , Adipose Tissue/cytology , Animals , Cell Differentiation , Cell Proliferation , Ganciclovir/administration & dosage , Genes, Transgenic, Suicide , Genetic Vectors , Humans , Lentivirus/genetics , Luminescent Measurements , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays/methods
11.
Mol Imaging Biol ; 11(3): 144-58, 2009.
Article in English | MEDLINE | ID: mdl-19048345

ABSTRACT

PURPOSE: Protein phosphorylation mediated by protein kinases controls numerous cellular processes. A genetically encoded, generalizable split firefly luciferase (FL)-assisted complementation system was developed for noninvasive monitoring phosphorylation events and efficacies of kinase inhibitors in cell culture and in small living subjects by optical bioluminescence imaging. PROCEDURES: An Akt sensor (AST) was constructed to monitor Akt phosphorylation and the effect of different PI-3K and Akt inhibitors. Specificity of AST was determined using a non-phosphorylable mutant sensor containing an alanine substitution (ASA). RESULTS: The PI-3K inhibitor LY294002 and Akt kinase inhibitor perifosine led to temporal- and dose-dependent increases in complemented FL activities in 293T human kidney cancer cells stably expressing AST (293T/AST) but not in 293T/ASA cells. Inhibition of endogenous Akt phosphorylation and kinase activities by perifosine also correlated with increase in complemented FL activities in 293T/AST cells but not in 293T/ASA cells. Treatment of nude mice bearing 293T/AST xenografts with perifosine led to a 2-fold increase in complemented FL activities compared to that of 293T/ASA xenografts. Our system was used to screen a small chemical library for novel modulators of Akt kinase activity. CONCLUSION: This generalizable approach for noninvasive monitoring of phosphorylation events will accelerate the discovery and validation of novel kinase inhibitors and modulators of phosphorylation events.


Subject(s)
Drug Discovery/methods , Molecular Probe Techniques , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Genetic Complementation Test , Humans , Luciferases, Firefly/genetics , Luminescence , Mice , Molecular Probes , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics
12.
Nano Lett ; 8(9): 2800-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18683988

ABSTRACT

An optimized noninvasive Raman microscope was used to evaluate tumor targeting and localization of single walled carbon nanotubes (SWNTs) in mice. Raman images were acquired in two groups of tumor-bearing mice. The control group received plain-SWNTs, whereas the experimental group received tumor targeting RGD-SWNTs intravenously. Raman imaging commenced over the next 72 h and revealed increased accumulation of RGD-SWNTs in tumor ( p < 0.05) as opposed to plain-SWNTs. These results support the development of a new preclinical Raman imager.


Subject(s)
Nanotubes, Carbon , Neoplasms, Experimental/therapy , Spectrum Analysis, Raman/methods , Animals , Mice
13.
Proc Natl Acad Sci U S A ; 105(15): 5844-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18378895

ABSTRACT

Molecular imaging of living subjects continues to rapidly evolve with bioluminescence and fluorescence strategies, in particular being frequently used for small-animal models. This article presents noninvasive deep-tissue molecular images in a living subject with the use of Raman spectroscopy. We describe a strategy for small-animal optical imaging based on Raman spectroscopy and Raman nanoparticles. Surface-enhanced Raman scattering nanoparticles and single-wall carbon nanotubes were used to demonstrate whole-body Raman imaging, nanoparticle pharmacokinetics, multiplexing, and in vivo tumor targeting, using an imaging system adapted for small-animal Raman imaging. The imaging modality reported here holds significant potential as a strategy for biomedical imaging of living subjects.


Subject(s)
Diagnostic Imaging/methods , Spectrum Analysis, Raman/methods , Animals , Liver/cytology , Liver/metabolism , Liver/pathology , Mice , Nanoparticles/administration & dosage , Phantoms, Imaging
14.
IEEE Trans Med Imaging ; 27(1): 58-63, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18270062

ABSTRACT

We present a phantom study to evaluate the performance of the eXplore Optix (Advanced Research Technologies-GE Healthcare), the first commercially available time-domain tomography system for small animal fluorescence imaging, and compare its capabilities with the widely used IVIS 200 (Xenogen Corporation-Caliper) continuous wave planar imaging system. The eXplore Optix, based on point-wise illumination and collection scheme, is found to be a log order more sensitive with significantly higher detection depth and spatial resolution as compared with the wide-area illumination IVIS 200 under the conditions tested. A time-resolved detection system allows the eXplore Optix to measure the arrival time distribution of fluorescence photons. This enables fluorescence lifetime measurement, absorption mapping, and estimation of fluorescent inclusion depth, which in turn is used by a reconstruction algorithm to calculate the volumetric distribution of the fluorophore concentration. An increased acquisition time and lack of ability to image multiple animals simultaneously are the main drawbacks of the eXplore Optix as compared with the IVIS 200.


Subject(s)
Image Enhancement/instrumentation , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/veterinary , Tomography, Optical/instrumentation , Tomography, Optical/veterinary , Animals , Equipment Design , Equipment Failure Analysis , Image Enhancement/methods , Microscopy, Fluorescence/methods , Reproducibility of Results , Sensitivity and Specificity , Tomography, Optical/methods
15.
AJNR Am J Neuroradiol ; 29(3): 409-18, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18272565

ABSTRACT

SUMMARY: In this second article, we review the various strategies and applications that make use of reporter genes for molecular imaging of the brain in living subjects. These approaches are emerging as valuable tools for monitoring gene expression in diverse applications in laboratory animals, including the study of gene-targeted and trafficking cells, gene therapies, transgenic animals, and more complex molecular interactions within the central nervous system. Further development of more sensitive and selective reporters, combined with improvements in detection technology, will consolidate the position of in vivo reporter gene imaging as a versatile technique for greater understanding of intracellular biologic processes and underlying molecular neuropathology and will potentially establish a future role in the clinical management of patients with neurologic diseases.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Diagnostic Imaging/trends , Forecasting , Gene Expression Profiling/trends , Genes, Reporter , Molecular Probe Techniques/trends , Animals , Humans , Neuroanatomy/trends , Research/trends
16.
Gene Ther ; 15(8): 583-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18305574

ABSTRACT

Effective treatment for recurrent, disseminated prostate cancer is notably limited. We have developed adenoviral vectors with a prostate-specific two-step transcriptional amplification (TSTA) system that would express therapeutic genes at a robust level to target metastatic disease. The TSTA system employs the prostate-specific antigen (PSA) promoter/enhancer to drive a potent synthetic activator, which in turn activates the expression of the therapeutic gene. In this study, we explored different configurations of this bipartite system and discovered that physical separation of the two TSTA components into E1 and E3 regions of adenovirus was able to enhance androgen regulation and cell-discriminatory expression. The TSTA vectors that express imaging reporter genes were assessed by noninvasive imaging technologies in animal models. The improved selectivity of the E1E3 configured vector was reflected in silenced ectopic expression in the lung. Significantly, the enhanced specificity of the E1E3 vector enabled the detection of lung metastasis of prostate cancer. An E1E3 TSTA vector that expresses the herpes simplex virus thymidine kinase gene can effectively direct positron emission tomography (PET) imaging of the tumor. The prostate-targeted gene delivery vectors with robust and cell-specific expression capability will advance the development of safe and effective imaging guided therapy for recurrent metastatic stages of prostate cancer.


Subject(s)
Adenoviridae/genetics , Gene Amplification , Genes, Reporter , Genetic Vectors/genetics , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/diagnostic imaging , Adenovirus E1 Proteins/genetics , Adenovirus E3 Proteins/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Gene Expression , Genetic Engineering , Humans , Lung Neoplasms/secondary , Male , Mice , Mice, SCID , Positron-Emission Tomography , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/pathology , Simplexvirus/enzymology , Thymidine Kinase/genetics , Transcription, Genetic
17.
AJNR Am J Neuroradiol ; 29(2): 229-34, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18024575

ABSTRACT

SUMMARY: In this first article, we review the basic principles of using reporter genes for molecular imaging of the brain in living subjects. This approach is emerging as a valuable tool for monitoring gene expression in diverse applications in laboratory animals, including the study of gene-targeted and trafficking cells, gene therapies, transgenic animals, and more complex molecular interactions within the central nervous system. Further development of more sensitive and selective reporters, combined with improvements in detection technology, will consolidate the position of in vivo reporter gene imaging as a versatile method for greater understanding of intracellular biologic processes and underlying molecular neuropathology and will potentially establish a future role in the clinical management of patients with neurologic diseases.


Subject(s)
Brain Diseases/diagnosis , Brain Diseases/metabolism , Brain/metabolism , Diagnostic Imaging/methods , Gene Targeting/methods , Genes, Reporter , Molecular Probe Techniques , Biomarkers/analysis , Brain/anatomy & histology , Humans
18.
Eur J Nucl Med Mol Imaging ; 35(3): 527-31, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17960376

ABSTRACT

PURPOSE: The aim of this study was to evaluate the potential of (18)F-fluorothymidine (FLT) PET/CT for imaging pancreatic adenocarcinoma. METHODS: This was a pilot study of five patients (four males, one female) with newly diagnosed and previously untreated pancreatic adenocarcinoma. Patients underwent FLT PET/CT, (18)F-fluorodeoxyglucose (FDG) PET/CT, and contrast-enhanced CT scanning before treatment. The presence of cancer was confirmed by histopathological analysis at the time of scanning in all five patients. The degree of FLT and FDG uptake at the primary tumor site was assessed using visual interpretation and semi-quantitative SUV analyses. RESULTS: The primary tumor size ranged from 2.5 x 2.8 cm to 3.5 x 7.0 cm. The SUV of FLT uptake within the primary tumor ranged from 2.1 to 3.1. Using visual interpretation, the primary cancer could be detected from background activity in two of five patients (40%) on FLT PET/CT. By comparison, FDG uptake was higher in each patient with a SUV range of 3.4 to 10.8, and the primary cancer could be detected from background in all five patients (100%). CONCLUSIONS: In this pilot study of five patients with primary pancreatic adenocarcinoma, FLT PET/CT scanning showed poor lesion detectability and relatively low levels of radiotracer uptake in the primary tumor.


Subject(s)
Adenocarcinoma/diagnosis , Dideoxynucleosides , Pancreatic Neoplasms/diagnosis , Positron-Emission Tomography/methods , Subtraction Technique , Tomography, X-Ray Computed/methods , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Pilot Projects , Radiopharmaceuticals , Reproducibility of Results , Sensitivity and Specificity
19.
Mol Imaging Biol ; 9(5): 260-6, 2007.
Article in English | MEDLINE | ID: mdl-17610017

ABSTRACT

PURPOSE: To evaluate 2-deoxy-2-[F-18]fluoro-D-glucose (FDG) accumulation in human ovarian carcinoma cell lines compared with control tumor cell lines known to accumulate FDG. PROCEDURES: FDG accumulation assays were performed in 15 different ovarian carcinoma cell lines at 1, 2, and 3 hours after incubation with 1 microCi of FDG. Results were compared with FDG accumulation in six different control tumor cell lines. 2-deoxy-2-[F-18]fluoro-D-glucose accumulation was expressed as counts per minute (cpm) in cells and normalized to initial cpm in medium and total protein content of cell lysates. RESULTS: FDG accumulation in all 15 ovarian carcinoma cell lines was equal to or higher than 0.0005 +/- 8.6 10(-5) cpm in cells/cpm in medium/mug protein at all three different time points. In two ovarian carcinoma cell lines (ES-2, poorly differentiated clear cell carcinoma, and OVCAR-3, poorly differentiated papillary adenocarcinoma), FDG accumulation was not statistically, significantly different compared to the control cell line with the highest FDG accumulation (LS 174T human colorectal adenocarcinoma) at two or more time points (P > or = 0.07). In 2 of 15 (13%) ovarian carcinoma cell lines (OVCAR5 epithelial carcinoma and SKOV3 clear cell carcinoma), FDG accumulation was lower than that in the control cell line with the lowest FDG accumulation (HT-29 human colorectal adenocarcinoma) at one or more time points (P < 0.05). CONCLUSIONS: Most human ovarian carcinoma cell lines showed comparable FDG accumulations with control cell lines known to accumulate FDG. This study lays the foundations for further comparisons with other ovarian cancer cell lines and for other positron emission tomography tracers.


Subject(s)
Fluorine Radioisotopes , Fluorodeoxyglucose F18 , Ovarian Neoplasms/diagnostic imaging , Radiopharmaceuticals , Animals , Cell Line, Tumor , Female , Fluorine Radioisotopes/pharmacokinetics , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Ovarian Neoplasms/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Rats
20.
Mol Imaging Biol ; 9(1): 50-7, 2007.
Article in English | MEDLINE | ID: mdl-17051322

ABSTRACT

OBJECTIVES: 2-Deoxy-2-[F-18]fluoro-D-glucose (FDG)-positron emission tomography (PET)/computed tomography (CT) is widely available as a powerful imaging modality, combining the ability to detect active metabolic processes and their morphologic features in a single exam. The role of FDG-PET is proven in a variety of cancers, including melanoma, but the estimates of sensitivity and specificity are based in the majority of the published studies on dedicated PET, not PET/CT. Therefore, we were prompted to review our experience with FDG-PET/CT in the management of melanoma. METHODS: This is a retrospective study on 106 patients with melanoma (20-87 years old; average: 56.8 +/- 15.9), who had whole-body FDG-PET/CT at our institution from January 2003 to June 2005. Thirty-eight patients (35.9%) were women and 68 patients (64.1%) were men. Reinterpretation of the imaging studies for accuracy and data analysis from medical records were performed. RESULTS: All patients had the study for disease restaging. The primary tumor depth (Breslow's thickness) at initial diagnosis was available for 76 patients (71.7%) and ranged from 0.4 to 25 mm (average: 3.56 mm). The anatomic level of invasion in the skin (Clark's level) was determined for 70 patients (66%): 3, level II; 13, level III; 43, level IV; 11, level V. The administered dose of (18)F FDG ranged from 9.8 to 21.6 mCi (average: 15.4 +/- 1.8 mCi). FDG-PET/CT had a sensitivity of 89.3% [95% confidence interval (CI): 78.5-95] and a specificity of 88% (95% CI: 76.2-94.4) for melanoma detection. CONCLUSION: This study confirms the good results of FDG-PET/CT for residual/recurrent melanoma detection, as well as for distant metastases localization. PET/CT should be an integral part in evaluation of patients with high-risk melanoma, prior to selection of the most appropriate therapy.


Subject(s)
Fluorine Radioisotopes , Fluorodeoxyglucose F18 , Melanoma/diagnostic imaging , Positron-Emission Tomography/methods , Tomography, Emission-Computed/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Melanoma/diagnosis , Middle Aged , Radiography , Retrospective Studies , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...