Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
J Nucl Med ; 52(6): 986-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21622896

ABSTRACT

The chemokine receptor 4 (CXCR4) is overexpressed in several cancers and metastases and as such presents an enticing target for molecular imaging of metastases and metastatic potential of the primary tumor. CXCR4-based imaging agents could also be useful for diagnosis, staging, and therapeutic monitoring. Here we evaluated a positron-emitting monocyclam analog, (64)Cu-{N-[1,4,8,11-tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine} ((64)Cu-AMD3465), in subcutaneous U87 brain tumors and U87 tumors stably expressing CXCR4 (U87-stb-CXCR4) and in colon tumors (HT-29) using dynamic and whole-body PET supported by ex vivo biodistribution studies. Both dynamic and whole-body PET/CT studies show specific accumulation of radioactivity in U87-stb-CXCR4 tumors, with the percentage injected dose per gram reaching a maximum of 102.70 ± 20.80 at 60 min and tumor-to-muscle ratios reaching a maximum of 362.56 ± 153.51 at 90 min after injection of the radiotracer. Similar specificity was also observed in the HT-29 colon tumor model. Treatment with AMD3465 inhibited uptake of radioactivity by the tumors in both models. Our results show that (64)Cu-AMD3465 is capable of detecting lesions in a CXCR4-dependent fashion, with high target selectivity, and may offer a scaffold for the synthesis of clinically translatable agents.


Subject(s)
Brain Neoplasms/diagnostic imaging , Neoplasm Transplantation/diagnostic imaging , Pyridines , Radiopharmaceuticals , Receptors, CXCR4/biosynthesis , Animals , Cell Line, Tumor , Colonic Neoplasms/diagnostic imaging , Copper Radioisotopes , Female , Flow Cytometry , HT29 Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Positron-Emission Tomography , Pyridines/chemical synthesis , Pyridines/chemistry , Radioligand Assay , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Solubility , Tissue Distribution , Whole-Body Counting
3.
J Med Chem ; 53(14): 5333-41, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20568777

ABSTRACT

Gallium-68 is a generator-produced radionuclide for positron emission tomography (PET) that is being increasingly used for radiolabeling of tumor-targeting peptides. Compounds [(68)Ga]3 and [(68)Ga]6 are high-affinity urea-based inhibitors of the prostate-specific membrane antigen (PSMA) that were synthesized in decay-uncorrected yields ranging from 60% to 70% and radiochemical purities of more than 99%. Compound [(68)Ga]3 demonstrated 3.78 +/- 0.90% injected dose per gram of tissue (%ID/g) within PSMA+ PIP tumor at 30 min postinjection, while [(68)Ga]6 showed a 2 h PSMA+ PIP tumor uptake value of 3.29 +/- 0.77 %ID/g. Target (PSMA+ PIP) to nontarget (PSMA- flu) ratios were 4.6 and 18.3, respectively, at those time points. Both compounds delineated tumor clearly by small animal PET. The urea series of imaging agents for PSMA can be radiolabeled with (68)Ga, a cyclotron-free isotope useful for clinical PET studies, with maintenance of target specificity.


Subject(s)
Organometallic Compounds/chemical synthesis , Prostate-Specific Antigen/antagonists & inhibitors , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals/chemical synthesis , Animals , Cell Line, Tumor , Gallium Radioisotopes , Humans , Isotope Labeling , Male , Mice , Mice, SCID , Neoplasm Transplantation , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Tissue Distribution , Transplantation, Heterologous
4.
Bioorg Med Chem Lett ; 20(1): 392-7, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19897367

ABSTRACT

We report a strategy based on bioisosterism to improve the physicochemical properties of existing hydrophilic, urea-based GCPII inhibitors. Comprehensive structure-activity relationship studies of the P1' site of ZJ-43- and DCIBzL-based compounds identified several glutamate-free inhibitors with K(i) values below 20nM. Among them, compound 32d (K(i)=11nM) exhibited selective uptake in GCPII-expressing tumors by SPECT-CT imaging in mice. A novel conformational change of amino acids in the S1' pharmacophore pocket was observed in the X-ray crystal structure of GCPII complexed with 32d.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Lysine/analogs & derivatives , Radiopharmaceuticals/chemistry , Urea/analogs & derivatives , Animals , Binding Sites , Crystallography, X-Ray , Glutamate Carboxypeptidase II/metabolism , Lysine/chemical synthesis , Lysine/chemistry , Lysine/pharmacology , Mice , Radiopharmaceuticals/chemical synthesis , Structure-Activity Relationship , Tomography, Emission-Computed, Single-Photon , Urea/chemical synthesis , Urea/chemistry , Urea/pharmacology
5.
Cell Res ; 19(3): 370-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19114988

ABSTRACT

Two new types of lentiviral vectors expressing a reporter transgene encoding either firefly luciferase (fLuc) for bioluminescence imaging or the HSV1 thymidine kinase (HSV1-TK) for radiopharmaceutical-based imaging were constructed to monitor human embryonic stem cell (hESC) engraftment and proliferation in live mice after transplantation. The constitutive expression of either transgene did not alter the properties of hESCs in the culture. We next monitored the formation of teratomas in SCID mice to test (1) whether the gene-modified hESCs maintain their developmental pluripotency, and (2) whether sustained reporter gene expression allows noninvasive, whole-body imaging of hESC derivatives in a live mouse model. We observed teratoma formation from both types of gene-modified cells as well as wild-type hESCs 2-4 months after inoculation. Using an optical imaging system, bioluminescence from the fLuc-transduced hESCs was easily detected in mice bearing teratomas long before palpable tumors could be detected. To develop a noninvasive imaging method more readily translatable to the clinic, we also utilized HSV1-TK and its specific substrate, 1-(2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl)-5-[(125)I]iodouracil ([(125)I]FIAU), as a reporter/probe pair. After systemic administration, [(125)I]FIAU is phosphorylated only by the transgene-encoded HSV1-TK enzyme and retained within transduced (and transplanted) cells, allowing sensitive and quantitative imaging by single-photon emission computed tomography. Noninvasive imaging methods such as these may enable us to monitor the presence and distribution of transplanted human stem cells repetitively within live recipients over a long term through the expression of a reporter gene.


Subject(s)
Embryonic Stem Cells/cytology , Stem Cell Transplantation , Teratoma/pathology , Whole Body Imaging/methods , Animals , Cell Line , Disease Models, Animal , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Luciferases/metabolism , Mice , Thymidine Kinase/metabolism , Tomography, Emission-Computed, Single-Photon , Transduction, Genetic
6.
J Med Chem ; 51(24): 7933-43, 2008 Dec 25.
Article in English | MEDLINE | ID: mdl-19053825

ABSTRACT

To extend our development of new imaging agents targeting the prostate-specific membrane antigen (PSMA), we have used the versatile intermediate 2-[3-(5-amino-1-carboxy-pentyl)-ureido]-pentanedioic acid (Lys-C(O)-Glu), which allows ready incorporation of radiohalogens for single photon emission computed tomography (SPECT) and positron emission tomography (PET). We prepared 2-[3-[1-carboxy-5-(4-[(125)I]iodo-benzoylamino)-pentyl]-ureido]-pentanedioic acid ([(125)I]3), 2-[3-[1-carboxy-5-(4-[(18)F]fluoro-benzoylamino)-pentyl]-ureido]-pentanedioic acid ([(18)F]6), and 2-(3-[1-carboxy-5-[(5-[(125)I]iodo-pyridine-3-carbonyl)-amino]-pentyl]-ureido)-pentanedioic acid ([(125)I]8) in 65-80% (nondecay-corrected), 30-35% (decay corrected), and 59-75% (nondecay-corrected) radiochemical yields. Compound [(125)I]3 demonstrated 8.8 +/- 4.7% injected dose per gram (%ID/g) within PSMA(+) PC-3 PIP tumor at 30 min postinjection, which persisted, with clear delineation of the tumor by SPECT. Similar tumor uptake values at early time points were demonstrated for [(18)F]6 (using PET) and [(125)I]8. Because of the many radiohalogenated moieties that can be attached via the epsilon amino group, the intermediate Lys-C(O)-Glu is an attractive template upon which to develop new imaging agents for prostate cancer.


Subject(s)
Antigens, Surface/biosynthesis , Glutamate Carboxypeptidase II/biosynthesis , Prostatic Neoplasms/metabolism , Adenosine Triphosphate/chemistry , Animals , Antigens, Surface/chemistry , Binding Sites , Chemistry, Pharmaceutical/methods , Computational Biology/methods , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Drug Design , Glutamate Carboxypeptidase II/chemistry , Humans , Male , Models, Molecular , Molecular Conformation , Protein Binding , Radiopharmaceuticals/pharmacology
7.
Clin Cancer Res ; 14(19): 6116-24, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829490

ABSTRACT

PURPOSE: Animal models of breast cancer metastases that recapitulate the pattern of metastatic progression seen in patients are lacking; metastatic breast cancer models do not currently exist for evaluation of immune-mediated therapies. We have developed and characterized a preclinical model for the evaluation of immune-mediated metastatic breast cancer therapies. EXPERIMENTAL DESIGN: The NT2.5 mammary tumor cell line was injected into the left cardiac ventricle of immunotolerant transgenic neu-N mice and athymic nu/nu mice. Metastatic progression was monitored by bioluminescent, small-animal magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography/computed tomography imaging, and also by histopathology. Antigen expression in normal organs and tumor metastases was evaluated by Western blot analysis and flow cytometry. RESULTS: Left cardiac ventricle injection of NT2.5 cells yielded widespread metastases in bones, liver, and spleen. Three to four weeks after injection, mice exhibited hind limb paralysis and occasional abdominal enlargement. Bioluminescence imaging of metastatic progression was successful in nude mice but the bioluminescent cells were rejected in immunocompetent mice. Other imaging modalities allowed successful imaging of nonbioluminescent cells. Small-animal positron emission tomography imaging allowed visualization of disease, in vivo, in the bones and liver. Magnetic resonance imaging revealed initial dissemination of the tumor cells to the bone marrow. Small-animal single-photon emission computed tomography/computed tomography imaging identified metastatic bone lesions targeted by a radiolabeled antibody. CONCLUSION: The model closely recapitulates the pattern of metastatic spread in breast cancer. This immunotolerant metastatic model is a novel addition to existing breast cancer models and coupling the model with in vivo imaging greatly facilitates the evaluation of targeted immunotherapies of metastasis.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Receptor, ErbB-2/genetics , Animals , Cell Line, Tumor , Disease Progression , Female , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Rats , Receptor, ErbB-2/metabolism , Time Factors , Tomography, Emission-Computed, Single-Photon/methods
8.
Nat Med ; 14(10): 1118-22, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18776891

ABSTRACT

We investigated the possibility of using a pharmacologic agent to modulate viral gene expression to target radiotherapy to tumor tissue. In a mouse xenograft model, we had previously shown targeting of [(125)I]2'-fluoro-2'-deoxy-beta-D-5-iodouracil-arabinofuranoside ([(125)I]FIAU) to tumors engineered to express the Epstein-Barr virus thymidine kinase (EBV-TK). Here we extend those results to targeting of a therapeutic radiopharmaceutical [(131)I]FIAU to slow or stop tumor growth or to achieve tumor regression. These outcomes were achieved in xenografts with tumors that constitutively expressed the EBV-TK. With naturally infected EBV tumor cell lines (Burkitt's lymphoma and gastric carcinoma), activation of viral gene expression by pretreatment with bortezomib was required. Marked changes in tumor growth could also be achieved in naturally infected Kaposi's sarcoma herpesvirus tumors after pretreatment with bortezomib. Bortezomib-induced enzyme-targeted radiation therapy illustrates the possibility of pharmacologically modulating tumor gene expression to result in targeted radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Arabinofuranosyluracil/analogs & derivatives , Boronic Acids/pharmacology , Burkitt Lymphoma/radiotherapy , Iodine Radioisotopes/therapeutic use , Pyrazines/pharmacology , Radiopharmaceuticals/therapeutic use , Stomach Neoplasms/radiotherapy , Thymidine Kinase/metabolism , Animals , Arabinofuranosyluracil/therapeutic use , Bortezomib , Cell Line, Tumor , Herpesviridae Infections/radiotherapy , Herpesvirus 4, Human/enzymology , Humans , Male , Mice , Mice, SCID , Sarcoma, Kaposi/radiotherapy , Thymidine Kinase/genetics
9.
J Med Chem ; 51(15): 4504-17, 2008 Aug 14.
Article in English | MEDLINE | ID: mdl-18637669

ABSTRACT

The prostate-specific membrane antigen (PSMA) is increasingly recognized as a viable target for imaging and therapy of cancer. We prepared seven (99m)Tc/Re-labeled compounds by attaching known Tc/Re chelating agents to an amino-functionalized PSMA inhibitor (lys-NHCONH-glu) with or without a variable length linker moiety. K i values ranged from 0.17 to 199 nM. Ex vivo biodistribution and in vivo imaging demonstrated the degree of specific binding to engineered PSMA+ PC3 PIP tumors. PC3-PIP cells are derived from PC3 that have been transduced with the gene for PSMA. Despite demonstrating nearly the lowest PSMA inhibitory potency of this series, [(99m)Tc(CO)3( L1)] (+) ( L1 = (2-pyridylmethyl)2N(CH2) 4CH(CO2H)NHCO-(CH2) 6CO-NH-lys-NHCONH-glu) showed the highest, most selective PIP tumor uptake, at 7.9 +/- 4.0% injected dose per gram of tissue at 30 min postinjection. Radioactivity cleared from nontarget tissues to produce a PIP to flu (PSMA-PC3) ratio of 44:1 at 120 min postinjection. PSMA can accommodate the steric requirements of (99m)Tc/Re complexes within PSMA inhibitors, the best results achieved with a linker moiety between the epsilon amine of the urea lysine and the chelator.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Electrons , Glutamate Carboxypeptidase II/metabolism , Male , Mice , Molecular Structure , Neoplasm Transplantation , Protein Binding , Rhenium/chemistry , Structure-Activity Relationship , Technetium/chemistry , Urea/chemistry
10.
J Nucl Med ; 49(7): 1189-95, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552143

ABSTRACT

UNLABELLED: The sympathetic nervous system of the heart plays a key role in the pathophysiology of various cardiac diseases. Small-animal models are valuable for obtaining further insight into mechanisms of cardiac disease and therapy. To determine the translational potential of cardiac neuronal imaging from rodents to humans, we characterized the rat sympathetic nervous system using 3 radiotracers that reflect different subcellular mechanisms: (11)C-meta-hydroxyephedrine (HED), a tracer of neuronal transport showing stable uptake and no washout in healthy humans; (11)C-phenylephrine (PHEN), a tracer of vesicular leakage and intraneuronal metabolic degradation with initial uptake and subsequent washout in humans; and (11)C-epinephrine (EPI), a tracer of vesicular storage with stable uptake and no washout in humans. METHODS: We used a small-animal PET system to study healthy male Wistar rats at baseline, after desipramine (DMI) pretreatment (DMI block), and with DMI injection 15 min after tracer delivery (DMI chase). The rats were kept under general isoflurane anesthesia while dynamic emission scans of the heart were recorded for 60 min after radiotracer injection. A myocardial retention index was determined by normalizing uptake at 40 min to the integral under the arterial input curve. Washout rates were determined by monoexponential fitting of myocardial time-activity curves. RESULTS: At baseline, HED showed high myocardial uptake and sustained retention, EPI showed moderate uptake and significant biphasic washout, and PHEN showed moderate uptake and monoexponential washout. The average (+/- SD) left ventricular retention index for HED, PHEN, and EPI was 7.38% +/- 0.82%/min, 3.43% +/- 0.45%/min, and 4.24% +/- 0.59%/min, respectively; the washout rate for HED, PHEN, and EPI was 0.13% +/- 0.23%/min, 1.13% +/- 0.35%/min, and 0.50% +/- 0.24%/min, respectively. The DMI chase resulted in increased washout only for HED. DMI block decreased myocardial uptake of all tracers by less than 90%. CONCLUSION: Kinetic profiles of HED in the rat myocardium were similar to those of HED in humans, suggesting comparable neuronal transport density. Unlike in humans, however, significant washout of EPI and faster washout of PHEN were encountered, consistent with high intraneuronal metabolic activity, high catecholamine turnover, and reduced vesicular storage. This evidence of increased neuronal activity in rodents has implications for translational studies of cardiac neuronal biology in humans.


Subject(s)
Ephedrine/analogs & derivatives , Epinephrine/pharmacokinetics , Myocardium/metabolism , Neurons/metabolism , Phenylephrine/pharmacokinetics , Sympathetic Nervous System/metabolism , Animals , Carbon Radioisotopes , Desipramine/pharmacology , Ephedrine/pharmacokinetics , Male , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar
11.
Clin Cancer Res ; 14(10): 3036-43, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483369

ABSTRACT

PURPOSE: Previously, we showed successful imaging of xenografts that express the prostate-specific membrane antigen (PSMA) using small-animal positron emission tomography (PET) and the radiolabeled PSMA inhibitor N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-S-[11C]methyl-l-cysteine. Herein, we extend that work by preparing and testing a PSMA inhibitor of the same class labeled with fluorine-18. EXPERIMENTAL DESIGN: N-[N-[(S)-1,3-Dicarboxypropyl]carbamoyl]-4-[18F]fluorobenzyl-l-cysteine ([18F]DCFBC) was prepared by reacting 4-[18F]fluorobenzyl bromide with the precursor (S)-2-[3-[(R)-1-carboxy-2-mercaptoethyl]ureido]-pentanedioic acid in ammonia-saturated methanol at 60 degrees C for 10 min followed by purification using C-18 reverse-phase semipreparative high-performance liquid chromatography. Severe combined immunodeficient mice bearing a s.c. PSMA+ PC-3 PIP tumor behind one shoulder and a PSMA(-) PC-3 FLU tumor behind the other shoulder were injected via the tail vein with either 1.85 MBq (50 microCi) of [18F]DCFBC for ex vivo biodistribution or 7.4 MBq (200 microCi) for imaging. For biodistribution, mice were sacrificed at 5, 15, 30, 60, and 120 min. Tumor, blood, and major organs were harvested and weighed, and radioactivity was counted. Imaging was done on the GE eXplore Vista small-animal PET scanner by collecting 12 consecutive 10-min frames. RESULTS: Radiochemical yield for [18F]DCFBC averaged 16 +/- 6% (n = 8) from 4-[18F]fluorobenzyl bromide. Specific radioactivities ranged from 13 to 133 GBq/micromol (350-3,600 Ci/mmol) with an average of 52 GBq/micromol (1,392 Ci/mmol; n = 6). Biodistribution and imaging studies showed high uptake of [18F]DCFBC in the PIP tumors with little to no uptake in FLU tumors. High radiopharmaceutical uptake was also seen in kidneys and bladder; however, washout of radioactivity from these organs was faster than from the PIP tumors. The maximum PIP tumor uptake was 8.16 +/- 2.55% injected dose per gram, achieved at 60 min after injection, which decreased to 4.69 +/- 0.89 at 120 min. The PIP tumor to muscle ratio was 20 at 120 min after injection. Based on the mouse biodistribution, the dose-limiting organ is the kidneys (human estimated absorbed dose: 0.05 mGy/MBq; 0.2 rad/mCi). CONCLUSION: [18F]DCFBC localizes to PSMA+-expressing tumors in mice, permitting imaging by small-animal PET. This new radiopharmaceutical is an attractive candidate for further studies of PET imaging of prostate cancer.


Subject(s)
Cysteine/analogs & derivatives , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Animals , Cysteine/chemical synthesis , Cysteine/pharmacokinetics , Fluorine Radioisotopes/pharmacokinetics , Humans , Male , Mice , Prostate-Specific Antigen/metabolism
12.
Mol Imaging ; 6(2): 131-9, 2007.
Article in English | MEDLINE | ID: mdl-17445507

ABSTRACT

Global expression profiling of pancreatic cancers has identified two cell surface molecules, claudin 4 and prostate stem cell antigen (PSCA), as being overexpressed in the vast majority of cases. Two antibodies, anti-claudin 4 and anti-PSCA, were radiolabeled with iodine 125 ((125)I) for imaging pancreatic cancer xenografts in mice using gamma scintigraphy and single-photon emission computed tomography-computed tomography (SPECT-CT). Immunofluorescence staining of intact and permeabilized Colo357 human pancreatic cancer cells showed strong extracellular staining by both anti-PSCA and anti-claudin 4. Biodistribution studies in claudin 4 and PSCA-expressing Colo357 and PANC-1 subcutaneous xenograft models in mice showed that [(125)I]anti-claudin 4 tumor to muscle ratio uptake was 4.3 in Colo357 at 6 days postinjection and 6.3 in PANC-1 xenografts at 4 days postinjection. Biodistribution of [(125)I]anti-PSCA showed tumor to muscle ratio uptake of 4.9 in Colo357 at 6 days postinjection. Planar gamma scintigraphic imaging in Colo357 xenograft-bearing mice showed clear tumor uptake of [(125)I]anti-claudin 4 by 24 hours postinjection and by 48 hours postinjection for [(125)I]anti-PSCA. SPECT-CT imaging with [(125)I]anti-claudin 4 and [(125)I]anti-PSCA in an L3.6PL orthotopic xenograft model showed strong tumor and spleen uptake at 5 days postinjection. Both anti-claudin 4 and anti-PSCA demonstrate promise as radiodiagnostic and possibly radiotherapeutic agents for human pancreatic cancers.


Subject(s)
Antigens, Neoplasm/analysis , Membrane Glycoproteins/analysis , Membrane Proteins/analysis , Neoplasm Proteins/analysis , Pancreatic Neoplasms/diagnosis , Radioimmunodetection/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Claudin-4 , Epitopes/immunology , Fluorescent Antibody Technique , GPI-Linked Proteins , Humans , Iodine Radioisotopes/chemistry , Membrane Glycoproteins/immunology , Membrane Proteins/immunology , Mice , Neoplasm Proteins/immunology , Neoplasm Transplantation
13.
Clin Cancer Res ; 13(5): 1453-8, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332288

ABSTRACT

PURPOSE: EBV and other herpesviruses are associated with a variety of malignancies. The EBV thymidine kinase (TK) is either not expressed or is expressed at very low levels in EBV-associated tumors. However, EBV-TK expression can be induced in vitro with several chemotherapeutic agents that promote viral lytic induction. The goal of this study is to image EBV-associated tumors by induction of viral TK expression with radiolabeled 2'-fluoro-2'-deoxy-beta-D-5-iodouracil-arabinofuranoside (FIAU). EXPERIMENTAL DESIGN: Immunoblot, luciferase reporter assay, and in vitro assay with [(14)C]FIAU were used to show the effects of bortezomib on the induction of lytic gene expression of EBV-associated tumor cells. In vivo imaging and ex vivo biodistribution studies with [(125)I]FIAU on EBV-associated tumors were done to visualize and confirm, respectively, the EBV(+) tumor-specific effects of bortezomib. RESULTS: In vitro assays with [(14)C]FIAU and ex vivo biodistribution studies with [(125)I]FIAU showed that uptake and retention of radiolabeled FIAU was specific for cells that express EBV-TK. Planar gamma imaging of EBV(+) Burkitt's lymphoma xenografts in severe combined immunodeficient mice showed [(125)I]FIAU localization within tumors following treatment with bortezomib. CONCLUSIONS: These results indicate the feasibility of imaging chemotherapy-mediated viral lytic induction by radiopharmaceutical-based techniques such as single photon emission computed tomography and positron emission tomography.


Subject(s)
Arabinofuranosyluracil/analogs & derivatives , Burkitt Lymphoma/diagnostic imaging , Diagnostic Imaging , Genes, Viral , Radiopharmaceuticals , Thymidine Kinase/genetics , Animals , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Bortezomib , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/virology , Gene Expression , Gene Transfer Techniques , Herpesvirus 4, Human/enzymology , Herpesvirus 4, Human/genetics , Humans , Immunoblotting , Mice , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Oncogenic Viruses/enzymology , Oncogenic Viruses/genetics , Pyrazines/therapeutic use , Radionuclide Imaging , Reverse Transcriptase Polymerase Chain Reaction , Thymidine Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...