Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 36(6): 1394-1410.e12, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38838644

RESUMEN

A vexing problem in mitochondrial medicine is our limited capacity to evaluate the extent of brain disease in vivo. This limitation has hindered our understanding of the mechanisms that underlie the imaging phenotype in the brain of patients with mitochondrial diseases and our capacity to identify new biomarkers and therapeutic targets. Using comprehensive imaging, we analyzed the metabolic network that drives the brain structural and metabolic features of a mouse model of pyruvate dehydrogenase deficiency (PDHD). As the disease progressed in this animal, in vivo brain glucose uptake and glycolysis increased. Propionate served as a major anaplerotic substrate, predominantly metabolized by glial cells. A combination of propionate and a ketogenic diet extended lifespan, improved neuropathology, and ameliorated motor deficits in these animals. Together, intermediary metabolism is quite distinct in the PDHD brain-it plays a key role in the imaging phenotype, and it may uncover new treatments for this condition.


Asunto(s)
Encéfalo , Glucosa , Propionatos , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa , Animales , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/metabolismo , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagen , Glucosa/metabolismo , Propionatos/metabolismo , Ratones , Dieta Cetogénica , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino , Glucólisis
2.
ACS Nano ; 17(7): 6178-6192, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36971591

RESUMEN

Macrophages comprise a significant portion of the immune cell compartment within tumors and are known contributors to tumor pathology; however, cancer immunotherapies targeting these cells are not clinically available. The iron oxide nanoparticle, ferumoxytol (FH), may be utilized as a nanophore for drug delivery to tumor-associated macrophages. We have demonstrated that a vaccine adjuvant, monophosphoryl lipid A (MPLA), can be stably captured within the carbohydrate shell of ferumoxytol without chemical modification of either the drug or the nanophore. This drug-nanoparticle combination (FH-MPLA) activated macrophages to an antitumorigenic phenotype at clinically relevant concentrations. In the immunotherapy-resistant B16-F10 model of murine melanoma, FH-MPLA treatment induced tumor necrosis and regression in combination with agonistic α-CD40 monoclonal antibody therapy. FH-MPLA, composed of clinically approved nanoparticle and drug payload, represents a potential cancer immunotherapy with translational relevance. FH-MPLA may be useful as an adjunctive therapy to existing antibody-based cancer immunotherapies which target only lymphocytic cells, reshaping the tumor immune environment.


Asunto(s)
Anticuerpos Monoclonales , Melanoma , Ratones , Animales , Preparaciones Farmacéuticas , Anticuerpos Monoclonales/farmacología , Óxido Ferrosoférrico , Inmunoterapia , Melanoma/tratamiento farmacológico
3.
Sci Rep ; 10(1): 13826, 2020 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-32778663

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Sci Rep ; 10(1): 10379, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587315

RESUMEN

The emergence of PET probes for amyloid plaques and neurofibrillary tangles, hallmarks of Alzheimer disease (AD), enables monitoring of pathology in AD mouse models. However, small-animal PET imaging is limited by coarse spatial resolution. We have installed a custom-fabricated PET insert into our small-animal MRI instrument and used PET/MRI hybrid imaging to define regions of amyloid vulnerability in 5xFAD mice. We compared fluorine-18 [18F]-Florbetapir uptake in the 5xFAD brain by dedicated small-animal PET/MRI and PET/CT to validate the quantitative measurement of PET/MRI. Next, we used PET/MRI to define uptake in six brain regions. As expected, uptake was comparable to wild-type in the cerebellum and elevated in the cortex and hippocampus, regions implicated in AD. Interestingly, uptake was highest in the thalamus, a region often overlooked in AD studies. Development of small-animal PET/MRI enables tracking of brain region-specific pathology in mouse models, which may prove invaluable to understanding AD progression and therapeutic development.


Asunto(s)
Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Imagen por Resonancia Magnética/métodos , Placa Amiloide/patología , Tomografía de Emisión de Positrones/métodos , Tálamo/patología , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Animales , Radioisótopos de Flúor/metabolismo , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Placa Amiloide/diagnóstico por imagen , Placa Amiloide/metabolismo , Radiofármacos/metabolismo , Tálamo/diagnóstico por imagen , Tálamo/metabolismo
5.
Clin Cancer Res ; 26(1): 147-158, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515460

RESUMEN

PURPOSE: Small-molecule inhibitors have revolutionized treatment of certain genomically defined solid cancers. Despite breakthroughs in treating systemic disease, central nervous system (CNS) metastatic progression is common, and advancements in treating CNS malignancies remain sparse. By improving drug penetration across a variably permeable blood-brain barrier and diffusion across intratumoral compartments, more uniform delivery and distribution can be achieved to enhance efficacy. EXPERIMENTAL DESIGN: Ultrasmall fluorescent core-shell silica nanoparticles, Cornell prime dots (C' dots), were functionalized with αv integrin-binding (cRGD), or nontargeting (cRAD) peptides, and PET labels (124I, 89Zr) to investigate the utility of dual-modality cRGD-C' dots for enhancing accumulation, distribution, and retention (ADR) in a genetically engineered mouse model of glioblastoma (mGBM). mGBMs were systemically treated with 124I-cRGD- or 124I-cRAD-C' dots and sacrificed at 3 and 96 hours, with concurrent intravital injections of FITC-dextran for mapping blood-brain barrier breakdown and the nuclear stain Hoechst. We further assessed target inhibition and ADR following attachment of dasatinib, creating nanoparticle-drug conjugates (Das-NDCs). Imaging findings were confirmed with ex vivo autoradiography, fluorescence microscopy, and p-S6RP IHC. RESULTS: Improvements in brain tumor delivery and penetration, as well as enhancement in the ADR, were observed following administration of integrin-targeted C' dots, as compared with a nontargeted control. Furthermore, attachment of the small-molecule inhibitor, dasatinib, led to its successful drug delivery throughout mGBM, demonstrated by downstream pathway inhibition. CONCLUSIONS: These results demonstrate that highly engineered C' dots are promising drug delivery vehicles capable of navigating the complex physiologic barriers observed in a clinically relevant brain tumor model.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dasatinib/farmacología , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico , Nanopartículas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Dióxido de Silicio/química , Animales , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Dasatinib/química , Modelos Animales de Enfermedad , Glioblastoma/patología , Radioisótopos de Yodo/química , Ratones , Nanopartículas/química , Clasificación del Tumor , Oligopéptidos/química , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/química , Radioisótopos/química , Circonio/química
6.
Nat Nanotechnol ; 14(6): 616-622, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30911166

RESUMEN

Acute myeloid leukaemia is a fatal disease for most patients. We have found that ferumoxytol (Feraheme), an FDA-approved iron oxide nanoparticle for iron deficiency treatment, demonstrates an anti-leukaemia effect in vitro and in vivo. Using leukaemia cell lines and primary acute myeloid leukaemia patient samples, we show that low expression of the iron exporter ferroportin results in a susceptibility of these cells via an increase in intracellular iron from ferumoxytol. The reactive oxygen species produced by free ferrous iron lead to increased oxidative stress and cell death. Ferumoxytol treatment results in a significant reduction of disease burden in a murine leukaemia model and patient-derived xenotransplants bearing leukaemia cells with low ferroportin expression. Our findings show how a clinical nanoparticle previously considered largely biologically inert could be rapidly incorporated into clinical trials for patients with leukaemia with low ferroportin levels.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Óxido Ferrosoférrico , Leucemia Mieloide Aguda , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales , Animales , Línea Celular Tumoral , Aprobación de Drogas , Óxido Ferrosoférrico/farmacocinética , Óxido Ferrosoférrico/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Especies Reactivas de Oxígeno/metabolismo , Estados Unidos , United States Food and Drug Administration , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Pharm ; 15(6): 2133-2141, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29684277

RESUMEN

Antibodies labeled with positron-emitting isotopes have been used for tumor detection, predicting which patients may respond to tumor antigen-directed therapy, and assessing pharmacodynamic effects of drug interventions. Prolactin receptor (PRLR) is overexpressed in breast and prostate cancers and is a new target for cancer therapy. We evaluated REGN2878, an anti-PRLR monoclonal antibody, as an immunoPET reagent. REGN2878 was labeled with Zr-89 after conjugation with desferrioxamine B or labeled with I-131/I-124. In vitro determination of the half-maximal inhibitory concentration (IC50) of parental REGN2878, DFO-REGN2878, and iodinated REGN2878 was performed by examining the effect of the increasing amounts of these on uptake of trace-labeled I-131 REGN2878. REGN1932, a non-PRLR binding antibody, was used as a control. Imaging and biodistribution studies were performed in mice bearing tumor xenografts with various expression levels of PRLR, including MCF-7, transfected MCF-7/PRLR, PC3, and transfected PC3/PRLR and T4D7v11 cell lines. The specificity of uptake in tumors was evaluated by comparing Zr-89 REGN2878 and REGN1932, and in vivo competition compared Zr-89 REGN2878 uptake in tumor xenografts with and without prior injection of 2 mg of nonradioactive REGN2878. The competition binding assay of DFO-REGN2878 at ratios of 3.53-5.77 DFO per antibody showed IC50 values of 0.4917 and 0.7136 nM, respectively, compared to 0.3455 nM for parental REGN2878 and 0.3343 nM for I-124 REGN2878. Imaging and biodistribution studies showed excellent targeting of Zr-89 REGN2878 in PRLR-positive xenografts at delayed times of 189 h (presented as mean ± 1 SD, percent injected activity per mL (%IA/mL) 74.6 ± 33.8%IA/mL). In contrast, MCF-7/PRLR tumor xenografts showed a low uptake (7.0 ± 2.3%IA/mL) of control Zr-89 REGN1932 and a very low uptake and rapid clearance of I-124 REGN2878 (1.4 ± 0.6%IA/mL). Zr-89 REGN2878 has excellent antigen-specific targeting in various PRLR tumor xenograft models. We estimated, using image-based kinetic modeling, that PRLR antigen has a very rapid in vivo turnover half-life of ∼14 min from the cell membrane. Despite relatively modest estimated tumor PRLR expression numbers, PRLR-expressing cells have shown final retention of the Zr-89 REGN2878 antibody, with an uptake that appeared to be related to PRLR expression. This reagent has the potential to be used in clinical trials targeting PRLR.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Inmunoconjugados/administración & dosificación , Neoplasias/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radiofármacos/administración & dosificación , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Ratones , Ratones Desnudos , Imagen Molecular/métodos , Neoplasias/patología , Radiofármacos/química , Radiofármacos/inmunología , Radiofármacos/farmacocinética , Receptores de Prolactina/inmunología , Receptores de Prolactina/metabolismo , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Imaging Biol ; 19(5): 754-761, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28194631

RESUMEN

PURPOSE: The folate receptor (FR) has emerged as an interesting diagnostic and therapeutic drug target with many potential applications in oncologic and inflammatory disorders. It was therefore the aim of this study to develop a folate-derived Ga-68-based positron emission tomography (PET) imaging tracer that is straightforward to radiolabel and could be broadly used in clinical studies. We validated its target binding affinity and specificity and compared it to [99mTc]EC20, the folate single-photon emission computed tomography (SPECT) imaging tracer that has been most extensively studied clinically so far. PROCEDURES: The new folic acid-derived PET imaging agent is linked via a polyethyleneglycol linker to the chelator 1,4,7-triazacyclononane-1,4,7-trisacetic acid (NOTA). This new compound, NOTA-folate, was labeled with gallium-68. We tested the probe's stability in human plasma and its selectivity in vitro, using the FR-positive KB cell line as well as the FR-negative A549 cell line. The pharmacokinetic profile of [68Ga]NOTA-folate was evaluated in FR-positive KB mouse xenografts. Following intravenous injection of [68Ga]NOTA-folate (383 ± 53 µCi), PET/computed tomography (CT) imaging studies as well as biodistribution studies were performed using KB tumor-bearing mice (n = 3). In vitro as well as in vivo studies were performed in parallel with the SPECT imaging tracer [99mTc]EC20. RESULTS: In comparison to [99mTc]EC20 (radiochemical yield (RCY) = 82.0 ± 2.9 %, 91.8 ± 2.0 % purity), similar radiochemical yield (87.2 ± 6.9 %) and radiochemical purity (95.6 ± 1.8 %) could be achieved for [68Ga]NOTA-folate. For both tracers, we observed high affinity for FR-positive cells in vitro and high plasma stability. In PET/CT and biodistribution studies, [68Ga]NOTA-folate appeared to display slightly superior in vivo performance in comparison to [99mTc]EC20. In detail, 68Ga-NOTA-folate showed very good tumor uptake and retention (6.6 ± 1.1 %ID/g), relatively low kidney uptake (21.7 ± 1.1 %ID/g), and very low liver uptake (0.38 ± 0.08 %ID/g). In vivo blocking studies using a fivefold excess of EC20 reduced the tumor uptake to 2.5 ± 0.7 %ID/g, confirming receptor specific binding of [68Ga]NOTA-folate in vivo. CONCLUSION: We validated a new Ga-68 folate-based PET imaging agent with excellent pharmacokinetics and tumor uptake. Based on a head-to-head comparison between both tracers, [68Ga]NOTA-folate is a suitable imaging probe for the delineation of FR-positive tumors and a promising candidate for clinical translation.


Asunto(s)
Ácido Fólico/química , Radioisótopos de Galio/química , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones Desnudos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Unión Proteica , Suero/metabolismo , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único
9.
Proc Natl Acad Sci U S A ; 113(44): E6731-E6740, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27791119

RESUMEN

Immunological complexity in atherosclerosis warrants targeted treatment of specific inflammatory cells that aggravate the disease. With the initiation of large phase III trials investigating immunomodulatory drugs for atherosclerosis, cardiovascular disease treatment enters a new era. We here propose a radically different approach: implementing and evaluating in vivo a combinatorial library of nanoparticles with distinct physiochemical properties and differential immune cell specificities. The library's nanoparticles are based on endogenous high-density lipoprotein, which can preferentially deliver therapeutic compounds to pathological macrophages in atherosclerosis. Using the apolipoprotein E-deficient (Apoe-/-) mouse model of atherosclerosis, we quantitatively evaluated the library's immune cell specificity by combining immunological techniques and in vivo positron emission tomography imaging. Based on this screen, we formulated a liver X receptor agonist (GW3965) and abolished its liver toxicity while still preserving its therapeutic function. Screening the immune cell specificity of nanoparticles can be used to develop tailored therapies for atherosclerosis and other inflammatory diseases.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Inmunoterapia , Nanopartículas/química , Animales , Antiinflamatorios , Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Autorradiografía , Benzoatos/agonistas , Benzoatos/química , Bencilaminas/agonistas , Bencilaminas/química , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Lipoproteínas HDL/química , Lipoproteínas HDL/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Imagen Molecular , Nanomedicina , Nanopartículas/metabolismo , Tomografía de Emisión de Positrones/métodos , ARN Mensajero/metabolismo
10.
Bioconjug Chem ; 27(9): 2014-23, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27396694

RESUMEN

pH (low) insertion peptides (pHLIP peptides) target acidic extracellular environments in vivo due to pH-dependent cellular membrane insertion. Two variants (Var3 and Var7) and wild-type (WT) pHLIP peptides have shown promise for in vivo imaging of breast cancer. Two positron emitting radionuclides ((64)Cu and (18)F) were used to label the NOTA- and NO2A-derivatized Var3, Var7, and WT peptides for in vivo biodistribution studies in 4T1 orthotopic tumor-bearing BALB/c mice. All of the constructs were radiolabeled with (64)Cu or [(18)F]-AlF in good yield. The in vivo biodistribution of the 12 constructs in 4T1 orthotopic allografted female BALB/c mice indicated that NO2A-cysVar3, radiolabeled with either (18)F (4T1 uptake; 8.9 ± 1.7%ID/g at 4 h p.i.) or (64)Cu (4T1 uptake; 8.2 ± 0.9%ID/g at 4 h p.i. and 19.2 ± 1.8% ID/g at 24 h p.i.), shows the most promise for clinical translation. Additional studies to investigate other tumor models (melanoma, prostate, and brain tumor models) indicated the universality of tumor targeting of these tracers. From this study, future clinical translation will focus on (18)F- or (64)Cu-labeled NO2A-cysVar3.


Asunto(s)
Radioisótopos de Cobre , Espacio Extracelular/química , Radioisótopos de Flúor , Proteínas de la Membrana , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/farmacocinética , Ratones , Ratones Endogámicos BALB C , Trazadores Radiactivos , Relación Estructura-Actividad , Distribución Tisular
11.
Mol Imaging Biol ; 18(3): 386-92, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26493053

RESUMEN

PURPOSE: The current study presents [(18)F]PARPi as imaging agent for PARP1 expression. PROCEDURES: [(18)F]PARPi was generated by conjugating a 2H-phthalazin-1-one scaffold to 4-[(18)F]fluorobenzoic acid. Biochemical assays, optical in vivo competition, biodistribution analysis, positron emission tomography (PET)/X-ray computed tomography, and PET/magnetic resonance imaging studies were performed in subcutaneous and orthotopic mouse models of glioblastoma. RESULTS: [(18)F]PARPi shows suitable pharmacokinetic properties for brain tumor imaging (IC50 = 2.8 ± 1.1 nM; logPCHI = 2.15 ± 0.41; plasma-free fraction = 63.9 ± 12.6 %) and accumulates selectively in orthotopic brain tumor tissue. Tracer accumulation in subcutaneous brain tumors was 1.82 ± 0.21 %ID/g, whereas in healthy brain, the uptake was only 0.04 ± 0.01 %ID/g. CONCLUSIONS: [(18)F]PARPi is a selective PARP1 imaging agent that can be used to visualize glioblastoma in xenograft and orthotopic mouse models with high precision and good signal/noise ratios. It offers new opportunities to non-invasively image tumor growth and monitor interventions.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/enzimología , Glioblastoma/diagnóstico por imagen , Glioblastoma/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Autorradiografía , Proteínas Sanguíneas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Radioisótopos de Flúor , Glioblastoma/patología , Semivida , Humanos , Imagen por Resonancia Magnética , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/sangre , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacocinética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Distribución Tisular/efectos de los fármacos , Tomografía Computarizada por Rayos X
12.
Nano Lett ; 15(2): 864-8, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25559467

RESUMEN

Chelator-free nanoparticles for intrinsic radiolabeling are highly desirable for whole-body imaging and therapeutic applications. Several reports have successfully demonstrated the principle of intrinsic radiolabeling. However, the work done to date has suffered from much of the same specificity issues as conventional molecular chelators, insofar as there is no singular nanoparticle substrate that has proven effective in binding a wide library of radiosotopes. Here we present amorphous silica nanoparticles as general substrates for chelator-free radiolabeling and demonstrate their ability to bind six medically relevant isotopes of various oxidation states with high radiochemical yield. We provide strong evidence that the stability of the binding correlates with the hardness of the radioisotope, corroborating the proposed operating principle. Intrinsically labeled silica nanoparticles prepared by this approach demonstrate excellent in vivo stability and efficacy in lymph node imaging.


Asunto(s)
Nanopartículas/química , Radioisótopos/química , Dióxido de Silicio/química , Animales , Quelantes/química , Ratones , Ratones Desnudos , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Imagen Multimodal , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X
13.
Surgery ; 157(2): 331-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25616946

RESUMEN

BACKGROUND: Peritoneal carcinomatosis (PC) is a terminal progression of colorectal cancer (CRC). Poor response to cytoreductive operation and chemotherapy coupled with the inability to reliably track disease progression by the use of established diagnostic methods, make this a deadly disease. We examined the effectiveness of the oncolytic vaccinia virus GLV-1h153 as a therapeutic and diagnostic vehicle. We believe that viral expression of the human sodium iodide transporter (hNIS) provides both real-time monitoring of viral therapy and effective treatment of colorectal peritoneal carcinomatosis (CRPC). METHODS: Infectivity and cytotoxic effect of GLV-1h153 on CRC cell lines was assayed in vitro. Viral replication was examined by standard viral plaque assays. Orthotopic CRPC xenografts were generated in athymic nude mice and subsequently administered GLV-1h153 intraperitoneally. A decrease in tumor burden was assessed by mass. Orthotopic tumors were visualized by single-photon emission computed tomography/computed tomography after Iodine ((131)I) administration and by fluorescence optical imaging. RESULTS: GLV-1h153 infected and killed CRC cells in a time- and concentration-dependent manner. Viral replication demonstrated greater than a 2.35 log increase in titer over 4 days. Intraperitoneal treatment of orthotopic CRPC xenografts resulted in a substantial decrease in tumor burden. Infection of orthotopic xenografts was therapeutic and facilitated monitoring by (131)I-single-photon emission computed tomography/computed tomography via expression of hNIS in infected tissue. CONCLUSION: GLV-1h153 kills CRC in vitro effectively and decreases tumor burden in vivo. We demonstrate that GLV-1h153 can be used as an agent to provide accurate delineation of tumor burden in vivo. These findings indicate that GLV-1h153 has potential for use as a therapeutic and diagnostic agent in the treatment of CRPC.


Asunto(s)
Neoplasias Colorrectales/terapia , Viroterapia Oncolítica/métodos , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/terapia , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Radioisótopos de Yodo , Ratones , Ratones Desnudos , Siembra Neoplásica , Virus Oncolíticos/genética , Neoplasias Peritoneales/patología , Radiofármacos , Proteínas Recombinantes/genética , Simportadores/genética , Tomografía Computarizada de Emisión de Fotón Único , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Nucl Med ; 55(10): 1706-11, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25060196

RESUMEN

UNLABELLED: Advances in preclinical molecular imaging have generated new opportunities to noninvasively visualize the biodistribution and tumor targeting of nanoparticle therapeutics. Capitalizing on recent achievements in this area, we sought to develop an (89)Zr-based labeling strategy for liposomal nanoparticles that accumulate in tumors via passive targeting mechanisms. METHODS: (89)Zr-labeled liposomes were prepared using 2 different approaches: click labeling and surface chelation. Pharmacokinetic and biodistribution studies, as well as PET/CT imaging of the radiolabeled nanoparticles, were performed on a mouse model of breast cancer. In addition, a dual PET/optical probe was prepared by incorporation of a near-infrared fluorophore and tested in vivo by PET and near-infrared fluorescence imaging. RESULTS: The surface chelation approach proved to be superior in terms of radiochemical yield and stability, as well as in vivo performance. Accumulation of these liposomes in tumor peaked at 24 h after injection and was measured to be 13.7 ± 1.8 percentage injected dose per gram. The in vivo performance of this probe was not essentially perturbed by the incorporation of a near-infrared fluorophore. CONCLUSION: We have developed a highly modular and efficient strategy for the labeling of liposomal nanoparticles with (89)Zr. In xenograft and orthotopic mouse models of breast cancer, we demonstrated that the biodistribution of these nanoparticles can be visualized by PET imaging. In combination with a near-infrared dye, these liposomal nanoparticles can serve as bimodal PET/optical imaging agents. The liposomes target malignant growth, and their bimodal features may be useful for simultaneous PET and intraoperative imaging.


Asunto(s)
Colorantes Fluorescentes/química , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Circonio/química , Animales , Quelantes/química , Modelos Animales de Enfermedad , Membrana Dobles de Lípidos/química , Liposomas/química , Ratones , Nanotecnología/métodos , Radiofármacos/química , Tomografía Computarizada por Rayos X/métodos
15.
FASEB J ; 28(2): 676-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24186964

RESUMEN

We investigated the therapeutic efficacy of a replication-competent oncolytic vaccinia virus, GLV-1h153, carrying human sodium iodide symporter (hNIS), in combination with radioiodine in an orthotopic triple-negative breast cancer (TNBC) murine model. In vitro viral infection was confirmed by immunoblotting and radioiodine uptake assays. Orthotopic xenografts (MDA-MB-231 cells) received intratumoral injection of GLV-1h153 or PBS. One week after viral injection, xenografts were randomized into 4 treatment groups: GLV-1h153 alone, GLV-1h153 and (131)I (∼ 5 mCi), (131)I alone, or PBS, and followed for tumor growth. Kruskal-Wallis and Wilcoxon tests were performed for statistical analysis. Radiouptake assay showed a 178-fold increase of radioiodine uptake in hNIS-expressing infected cells compared with PBS control. Systemic (131)I-iodide in combination with GLV-1h153 resulted in a 6-fold increase in tumor regression (24 compared to 146 mm(3) for the virus-only treatment group; P<0.05; d 40). We demonstrated that a novel vaccinia virus, GLV-1h153, expresses hNIS, increases the expression of the symporter in TNBC cells, and serves both as a gene marker for noninvasive imaging of virus and as a vehicle for targeted radionuclide therapy with (131)I.


Asunto(s)
Radioisótopos de Yodo/uso terapéutico , Neoplasias de la Mama Triple Negativas/radioterapia , Neoplasias de la Mama Triple Negativas/terapia , Virus Vaccinia/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Neoplasias de la Mama Triple Negativas/metabolismo , Virus Vaccinia/genética
16.
Bioconjug Chem ; 24(11): 1784-9, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24180480

RESUMEN

Liposomes are attractive vehicles for the controlled release of drugs and cytotoxins and have a long-standing history in medical research and clinical practice. In addition to established therapeutic indications, liposomes have several favorable properties for molecular imaging, including high stability and the ability to be labeled with radioisotopes, as well as paramagnetic and fluorescent contrast agents. However, long circulation times and difficulties in creating targeted liposomes have proven challenges for imaging. In this study, we have addressed these limitations using a recently developed strategy for bioorthogonal conjugation, the reaction between tetrazines and trans-cyclooctenes. By coating radiolabeled liposomes with trans-cyclooctene and pretargeting with a tetrazine coupled to a targeted peptide, we were able to selectively enhance the retention of liposomes and bind them to tumor tissue in live animals. The rapid reaction between tetrazines and trans-cyclooctenes allowed imaging to be performed with the short-lived PET tracer (18)F, yielding signal-to-background activity ratios of 7:1. The covalent, bioorthogonally driven tumor-targeting of liposomes by in vivo click chemistry is promising and should be explored for more selective and rapid delivery of radiodiagnostics and radiotherapeutics, two classes of drugs which particularly benefit from fast clearance, low nonspecific binding, and the associated reduced toxicity to kidneys and bone marrow.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Radioisótopos de Flúor/metabolismo , Liposomas/química , Liposomas/metabolismo , Animales , Línea Celular Tumoral , Química Clic , Ciclooctanos/química , Humanos , Liposomas/farmacocinética , Ratones , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Tomografía de Emisión de Positrones , Tetrazoles/química
17.
Surgery ; 154(3): 486-95, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23890748

RESUMEN

BACKGROUND: The purpose of this work was to examine the ability of an oncolytic vaccinia virus expressing the human sodium iodine transporter (hNIS) to provide real time monitoring of viral therapy and effective treatment of malignant pleural mesothelioma (MPM). METHODS: Infectivity and cytotoxic effects of GLV-1h153 on mesothelioma cell lines of all histologic subtypes were assayed in vitro. Viral replication was examined by standard viral plaque assay. Orthotopic MPM xenografts were generated in athymic nude mice, treated with intrapleural GLV-1h153, and assessed for effect on tumor burden and survival. Orthotopic tumors were also imaged on single photon emission computed tomography (SPECT)/computed tomography (CT) after (131)I administration. RESULTS: GLV-1h153-infected and killed all cell lines in a time- and concentration-dependent manner. Viral replication demonstrated a >2.5-log increase in titer over 4 days. Intrapleural treatment of orthotopic MPM xenografts resulted in a significant decrease in tumor burden 1 week after treatment and an improvement in survival. Infection of orthotopic xenografts was both therapeutic and facilitated monitoring by (131)I-SPECT/CT via expression of hNIS in infected tissue. CONCLUSION: Our results suggest that GLV-1h153 may be a promising therapeutic agent for MPM and warrants further investigation.


Asunto(s)
Mesotelioma/terapia , Imagen Multimodal/métodos , Virus Oncolíticos/genética , Neoplasias Pleurales/terapia , Tomografía de Emisión de Positrones , Simportadores/genética , Tomografía Computarizada por Rayos X , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Mediciones Luminiscentes , Mesotelioma/diagnóstico por imagen , Mesotelioma/mortalidad , Ratones , Neoplasias Pleurales/diagnóstico por imagen , Neoplasias Pleurales/mortalidad , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Imaging Biol ; 15(6): 768-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23722880

RESUMEN

PURPOSE: This study aims to develop a molecular imaging strategy for response assessment of arginine deiminase (ADI) treatment in melanoma xenografts using 3'-[(18)F]fluoro-3'-deoxythymidine ([(18)F]-FLT) positron emission tomography (PET). PROCEDURES: F-FLT response to ADI therapy was studied in preclinical models of melanoma in vitro and in vivo. The molecular mechanism of response to ADI therapy was investigated, with a particular emphasis on biological pathways known to regulate (18)F-FLT metabolism. RESULTS: Proliferation of SK-MEL-28 melanoma tumors was potently inhibited by ADI treatment. However, no metabolic response was observed in FLT PET, presumably based on the known ADI-induced degradation of PTEN, followed by instability of the tumor suppressor p53 and a relative overexpression of thymidine kinase 1, the enzyme mainly responsible for intracellular FLT processing. CONCLUSION: The specific pharmacological properties of ADI preclude using (18)F-FLT to evaluate clinical response in melanoma and argue for further studies to explore the use of other clinically applicable PET tracers in ADI treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Didesoxinucleósidos/farmacocinética , Hidrolasas/uso terapéutico , Melanoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Didesoxinucleósidos/química , Hidrolasas/farmacología , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Melanoma/patología , Ratones , Transducción de Señal/efectos de los fármacos , Timidina Quinasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
AJR Am J Roentgenol ; 200(6): 1347-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23701074

RESUMEN

OBJECTIVE: Iodinated contrast agent for CT has a short half-life in the vasculature. As the field of interventional procedures expands, a more durable contrast agent would be highly useful. Our study investigated whether gold nanoparticles are feasible as a long-lasting vascular contrast agent for CT. MATERIALS AND METHODS: Gold nanoparticles were synthesized by a modified Turkevich method, coated with methoxy-polyethylene glycol-thiol chains, and compared with an iodine-based contrast agent used in mice. Contrast agents were imaged in tubes by CT at 40, 60, and 140 kVp and then were tested in vivo by tail vein injection. Nine mice received gold nanoparticles, two received iodine-based contrast agent, and one received saline. CT of mice was performed at 60 kVp immediately, 6 hours, and 24 hours after injection. RESULTS: In an isolated form in tubes, gold nanoparticles had greater radiographic density than did iodine-based contrast agent at 40 kVp and were comparable at the other CT voltages. In mice, gold nanoparticles provided bright contrast enhancement that enabled clear visualization of the abdominal aorta and renal arteries, which could not be distinguished without contrast agent. This persisted up to 24 hours, which was the last time point assessed. Contrast enhancement of the vasculature by iodine-based contrast agent was present immediately after injection but had disappeared by 6 hours. CONCLUSION: Gold nanoparticles can provide clear and durable contrast enhancement of the vasculature even at 24 hours. These findings merit further study of gold nanoparticles for their potential as a contrast agent in CT and CT-guided interventional procedures.


Asunto(s)
Medios de Contraste/química , Oro/química , Nanopartículas del Metal/química , Tomografía Computarizada por Rayos X , Animales , Gadolinio/química , Semivida , Ratones
20.
Mol Imaging ; 12(1): 67-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23348793

RESUMEN

Based on their inability to express argininosuccinate synthetase (ASS), some cancer entities feature the characteristic of L-arginine (Arg) auxotrophy. This inability to intrinsically generate Arg makes them applicable for arginine deiminase (ADI) treatment, an Arg-depleting drug. Arg is also used for the synthesis of endothelial nitric oxide (NO), which mainly confers vasodilatation but is also considered to have a major influence on tumor vascularization. The purpose of this study was to define changes in tumor vasculature in an ADI-treated melanoma xenograft mouse model using the blood pool agent AngioSense 750 and fluorescence molecular tomography (FMT). We used an ASS-negative melanoma xenograft mouse model and subjected it to weekly ADI treatment. Changes in tumor size were measured, and alterations in tumor vasculature were depicted by FMT and CD31 immunohistochemistry (IHC). On ADI treatment and effective antitumor therapy, we observed a drop in NO plasma levels and visualized changes in tumor vascularization with FMT and IHC. ADI treatment in melanoma xenografts has a tumor-reducing effect, which can be noninvasively imaged by quantifying tumor vascularization with FMT and IHC.


Asunto(s)
Hidrolasas/farmacología , Melanoma/irrigación sanguínea , Melanoma/tratamiento farmacológico , Imagen Óptica/métodos , Tomografía/métodos , Animales , Argininosuccinato Sintasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Imagen Molecular/métodos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Óxido Nítrico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA