Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Pharmacol Exp Ther ; 370(2): 288-298, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31152006

RESUMEN

In mouse pharmacokinetic (PK) studies, current standard methods often require large numbers of animals to support collection of blood samples serially over a defined time range. We have developed and validated a noninvasive fluorescence molecular tomography (FMT) heart imaging approach for blood PK quantification that uses small numbers of mice and has the advantage of repeated, longitudinal live imaging. This method was validated using a variety of near infrared (NIR) fluorescent-labeled molecules, ranging in size from 1.3 to 150 kDa, that were assessed by microplate blood assays as well as by noninvasive FMT 4000 imaging. Excellent agreement in kinetic profiles and calculated PK metrics was seen for the two methods, establishing the robustness of this noninvasive optical imaging approach. FMT heart imaging was further assessed in the challenging application of inulin-based glomerular filtration rate (GFR) measurement. After a single bolus injection of an NIR fluorescent-labeled inulin probe in small cohorts of mice (n = 5 per group), 2-minute heart scans (at 2, 6, 15, 30, and 45 minutes) were performed by FMT imaging. GFR was calculated using two-compartment PK modeling, determining an average rate of 240 ± 21 µl/min in normal mice, in agreement with published mouse GFR ranges. Validation of GFR assessment in unilaterally nephrectomized mice and cyclosporin A-treated mice both measured ∼50% decreases in GFR. Imaging results correlated well with ex vivo plasma microplate assays for inulin blood kinetics, and the decreases in GFR were accompanied by increases in plasma creatinine and blood urea nitrogen.


Asunto(s)
Sangre/metabolismo , Tasa de Filtración Glomerular/efectos de los fármacos , Corazón/diagnóstico por imagen , Imagen Óptica , Tomografía , Animales , Sangre/efectos de los fármacos , Creatinina/sangre , Femenino , Ratones , Nitrógeno/orina , Distribución Tisular
2.
Am J Physiol Renal Physiol ; 303(4): F593-603, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22674025

RESUMEN

The renin-angiotensin system (RAS) is well studied for its regulation of blood pressure and fluid homeostasis, as well as for increased activity associated with a variety of diseases and conditions, including cardiovascular disease, diabetes, and kidney disease. The enzyme renin cleaves angiotensinogen to form angiotensin I (ANG I), which is further cleaved by angiotensin-converting enzyme to produce ANG II. Although ANG II is the main effector molecule of the RAS, renin is the rate-limiting enzyme, thus playing a pivotal role in regulating RAS activity in hypertension and organ injury processes. Our objective was to develop a near-infrared fluorescent (NIRF) renin-imaging agent for noninvasive in vivo detection of renin activity as a measure of tissue RAS and in vitro plasma renin activity. We synthesized a renin-activatable agent, ReninSense 680 FAST (ReninSense), using a NIRF-quenched substrate derived from angiotensinogen that is cleaved specifically by purified mouse and rat renin enzymes to generate a fluorescent signal. This agent was assessed in vitro, in vivo, and ex vivo to detect and quantify increases in plasma and kidney renin activity in sodium-sensitive inbred C57BL/6 mice maintained on a low dietary sodium and diuretic regimen. Noninvasive in vivo fluorescence molecular tomographic imaging of the ReninSense signal in the kidney detected increased renin activity in the kidneys of hyperreninemic C57BL/6 mice. The agent also effectively detected renin activity in ex vivo kidneys, kidney tissue sections, and plasma samples. This approach could provide a new tool for assessing disorders linked to altered tissue and plasma renin activity and to monitor the efficacy of therapeutic treatments.


Asunto(s)
Colorantes Fluorescentes/farmacología , Péptidos/farmacología , Renina/sangre , Renina/metabolismo , Alimentación Animal/análisis , Animales , Catepsina D , Catepsina G , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Peptidil-Dipeptidasa A/metabolismo , Ratas , Sistema Renina-Angiotensina/fisiología , Sensibilidad y Especificidad , Sodio en la Dieta
3.
Bioorg Med Chem Lett ; 22(1): 653-7, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22079760

RESUMEN

A series of human carbonic anhydrase (hCA) IX inhibitors conjugated to various near-infrared fluorescent dyes was synthesized with the aim of imaging hypoxia-induced hCA IX expression in tumor cells in vitro, ex vivo and in vivo. The resulting compounds were profiled for inhibition of transmembrane hCA IX showing a range of potencies from 7.5 to 116 nM and up to 50-fold selectivity over the cytosolic form hCA II. Some of the compounds also showed inhibition selectivity for other transmembrane forms hCA XII and XIV as well. Compounds incubated in vitro with HeLa cells cultured under normoxic and hypoxic conditions detected upregulation of hCA IX under hypoxia by fluorescence microscopy. A pilot in vivo study in HT-29 tumor bearing mice showed significant accumulation of a fluorescent acetazolamide derivative in tumor tissue with little accumulation in other tissues. Approximately 10% of injected dose was non-invasively quantified in tumors by fluorescence molecular tomography (FMT), demonstrating the promise of these new compounds for quantitative imaging of hCA IX upregulation in live animals.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Anhidrasas Carbónicas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Neoplasias/patología , Sulfonamidas/farmacología , Animales , Anhidrasa Carbónica IX , Línea Celular Tumoral , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Colorantes Fluorescentes/farmacología , Células HeLa , Humanos , Hipoxia , Cinética , Ratones , Microscopía Fluorescente/métodos , Modelos Químicos , Trasplante de Neoplasias , Neoplasias/metabolismo , Tomografía Computarizada por Rayos X/métodos , Regulación hacia Arriba
4.
J Cardiovasc Pharmacol ; 57(4): 400-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21502925

RESUMEN

BACKGROUND: Antifibrinolytic therapy, such as the use of the serine protease inhibitor aprotinin, was a mainstay for hemostasis after cardiac surgery. However, aprotinin was empirically dosed, and although the pharmacological target was the inhibition of plasmin activity (PLact), this was never monitored, off-target effects occurred, and led to withdrawn from clinical use. The present study developed a validated fluorogenic microdialysis method to continuously measure PLact and tested the hypothesis that standardized clinical empirical aprotinin dosing would impart differential and regional effects on PLact. METHODS/RESULTS: Pigs (30 kg) were instrumented with microdialysis probes to continuously measure PLact in myocardial, kidney, and skeletal muscle compartments (deltoid) and then randomized to high-dose aprotinin administration (2 mKIU load/0.5 mKIU/hr infusion; n = 7), low-dose aprotinin administration (1 mKIU load/0.250 mKIU/hr infusion; n = 6). PLact was compared with time-matched vehicle (n = 4), and PLact was also measured in plasma by an in vitro fluorogenic method. Aprotinin suppressed PLact in the myocardium and kidney at both high and low doses, indicative that both doses exceeded a minimal concentration necessary for PLact inhibition. However, differential effects of aprotinin on PLact were observed in the skeletal muscle, indicative of different compartmentalization of aprotinin. CONCLUSIONS: Using a large animal model and a continuous method to monitor regional PLact, these unique results demonstrated that an empirical aprotinin dosing protocol causes maximal and rapid suppression in the myocardium and kidney and in turn would likely increase the probability of off-target effects and adverse events. Furthermore, this proof of principle study demonstrated that continuous monitoring of determinants of fibrinolysis might provide a novel approach for managing fibrinolytic therapy.


Asunto(s)
Aprotinina/farmacología , Fibrinolisina/metabolismo , Microdiálisis/métodos , Inhibidores de Serina Proteinasa/farmacología , Animales , Aprotinina/administración & dosificación , Aprotinina/efectos adversos , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes/química , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Inhibidores de Serina Proteinasa/administración & dosificación , Inhibidores de Serina Proteinasa/efectos adversos , Porcinos
5.
Arterioscler Thromb Vasc Biol ; 29(10): 1444-51, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19608968

RESUMEN

OBJECTIVE: Proteases are emerging biomarkers of inflammatory diseases. In atherosclerosis, these enzymes are often secreted by inflammatory macrophages, digest the extracellular matrix of the fibrous cap, and destabilize atheromata. Protease function can be monitored with protease activatable imaging probes and quantitated in vivo by fluorescence molecular tomography (FMT). To address 2 major constraints currently associated with imaging of murine atherosclerosis (lack of highly sensitive probes and absence of anatomic information), we compared protease sensors (PS) of variable size and pharmacokinetics and coregistered FMT datasets with computed tomography (FMT-CT). METHODS AND RESULTS: Coregistration of FMT and CT was achieved with a multimodal imaging cartridge containing fiducial markers detectable by both modalities. A high-resolution CT angiography protocol accurately localized fluorescence to the aortic root of atherosclerotic apoE(-/-) mice. To identify suitable sensors, we first modeled signal kinetics in-silico and then compared 3 probes with oligo-L-lysine cleavage sequences: PS-5, 5 nm in diameter containing 2 fluorochromes, PS-25, a 25-nm version with an elongated lysine chain and PS-40, a polymeric nanoparticle. Serial FMT-CT showed fastest kinetics for PS-5 but, surprisingly, highest fluorescence in lesions of the aortic root for PS-40. PS-40 robustly reported therapeutic effects of atorvastatin, corroborated by ex vivo imaging and qPCR for the model protease cathepsin B. CONCLUSIONS: FMT-CT is a robust and observer-independent tool for noninvasive assessment of inflammatory murine atherosclerosis. Reporter-containing nanomaterials may have unique advantages over small molecule agents for in vivo imaging.


Asunto(s)
Aterosclerosis/diagnóstico , Técnicas Biosensibles/métodos , Microscopía Fluorescente/métodos , Nanopartículas , Péptido Hidrolasas/metabolismo , Tomografía Computarizada por Rayos X/métodos , Animales , Apolipoproteínas E/fisiología , Aterosclerosis/enzimología , Citometría de Flujo , Semivida , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos
6.
PLoS One ; 12(8): e0182689, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28792505

RESUMEN

Physical measurement of tumor volume reduction is the most commonly used approach to assess tumor progression and treatment efficacy in mouse tumor models. However, it is relatively insensitive, and often requires long treatment courses to achieve gross physical tumor destruction. As alternatives, several non-invasive imaging methods such as bioluminescence imaging (BLI), fluorescence imaging (FLI) and positron emission tomography (PET) have been developed for more accurate measurement. As tumors have elevated glucose metabolism, 18F-fludeoxyglucose (18F-FDG) has become a sensitive PET imaging tracer for cancer detection, diagnosis, and efficacy assessment by measuring alterations in glucose metabolism. In particular, the ability of 18F-FDG imaging to detect drug-induced effects on tumor metabolism at a very early phase has dramatically improved the speed of decision-making regarding treatment efficacy. Here we demonstrated an approach with FLI that offers not only comparable performance to PET imaging, but also provides additional benefits, including ease of use, imaging throughput, probe stability, and the potential for multiplex imaging. In this report, we used sorafenib, a tyrosine kinase inhibitor clinically approved for cancer therapy, for treatment of a mouse tumor xenograft model. The drug is known to block several key signaling pathways involved in tumor metabolism. We first identified an appropriate sorafenib dose, 40 mg/kg (daily on days 0-4 and 7-10), that retained ultimate therapeutic efficacy yet provided a 2-3 day window post-treatment for imaging early, subtle metabolic changes prior to gross tumor regression. We then used 18F-FDG PET as the gold standard for assessing the effects of sorafenib treatment on tumor metabolism and compared this to results obtained by measurement of tumor size, tumor BLI, and tumor FLI changes. PET imaging showed ~55-60% inhibition of tumor uptake of 18F-FDG as early as days 2 and 3 post-treatment, without noticeable changes in tumor size. For comparison, two FLI probes, BombesinRSense™ 680 (BRS-680) and Transferrin-Vivo™ 750 (TfV-750), were assessed for their potential in metabolic imaging. Metabolically active cancer cells are known to have elevated bombesin and transferrin receptor levels on the surface. In excellent agreement with PET imaging, the BRS-680 imaging showed 40% and 79% inhibition on days 2 and 3, respectively, and the TfV-750 imaging showed 65% inhibition on day 3. In both cases, no significant reduction in tumor volume or BLI signal was observed during the first 3 days of treatment. These results suggest that metabolic FLI has potential preclinical application as an additional method for detecting drug-induced metabolic changes in tumors.


Asunto(s)
Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/tratamiento farmacológico , Imagen Óptica , Tomografía de Emisión de Positrones , Receptores de Bombesina/metabolismo , Receptores de Transferrina/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Fluorodesoxiglucosa F18 , Humanos , Ratones Transgénicos , Imagen Molecular , Trasplante de Neoplasias , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Radiofármacos , Distribución Aleatoria , Sorafenib , Resultado del Tratamiento , Carga Tumoral
7.
Cancer Res ; 62(21): 6146-51, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12414640

RESUMEN

The alpha(v)beta(3) integrin is expressed on proliferating endothelial cells such as those present in growing tumors, as well as on tumor cells of various origin. Tumor-induced angiogenesis can be blocked in vivo by antagonizing the alpha(v)beta(3) integrin with small peptides containing the Arg-Gly-Asp (RGD) amino acid sequence. This tripeptidic sequence, naturally present in extracellular matrix proteins, is the primary binding site of the alpha(v)beta(3) integrin. Because of selective expression of alpha(v)beta(3) integrin in tumors, radiolabeled RGD peptides are attractive candidates for alpha(v)beta(3) integrin targeting in tumors. We studied the in vivo behavior of the radiolabeled dimeric RGD peptide E-[c(RGDfK)](2) in the NIH:OVCAR-3 s.c. ovarian carcinoma xenograft model in BALB/c nude mice. Conjugation of the 1,4,7,10-tetraazadodecane-N,N',N",N"'-tetraacetic acid (DOTA) and hydrazinonicotinamide (HYNIC) chelators enabled efficient radiolabeling with (111)In/(90)Y and (99m)Tc, respectively. The radiolabeled peptide was rapidly excreted renally. Uptake in nontarget organs such as liver and spleen was considerable. Tumor uptake peaked at 7.5% injected dose (ID)/g ((111)In-DOTA-E-[c(RGDfK)](2)) or 6.0%ID/g ((99m)Tc-HYNIC-E-[c(RGDfK)](2)) at 2 and 1 h postinjection, respectively. Integrin alpha(v)beta(3) receptor binding specificity was demonstrated by reduced tumor uptake after injection of the scrambled control peptide (111)In-DOTA-E-[c(RDKfD)](2) (0.28%ID/g at 2 h p.i.) and after coinjection of excess nonradioactive (115)In-DOTA-E-[c(RGDfK)](2) (0.22%ID/g at 2 h p.i.). A single injection of (90)Y-DOTA-E-[c(RGDfK)](2) at the maximum-tolerated dose (37 MBq) in mice with small s.c. tumors caused a significant growth delay as compared with mice treated with 37 MBq (90)Y-labeled scrambled peptide or untreated mice (median survival of 54 versus 33.5 versus 19 days, respectively). In conclusion, the radiolabeled RGD peptides (111)In-DOTA-E-[c(RGDfK)](2) and (99m)Tc-HYNIC-E-[c(RGDfK)](2) demonstrated high and specific tumor uptake in a human tumor xenograft. Injection of (90)Y-DOTA-E-[c(RGDfK)](2) induced a significant delay in tumor growth. Potentially, these peptides can be used for peptide receptor radionuclide imaging as well as therapy.


Asunto(s)
Radioisótopos de Indio/uso terapéutico , Integrina alfaVbeta3/metabolismo , Oligopéptidos/metabolismo , Radiofármacos/metabolismo , Tecnecio/uso terapéutico , Radioisótopos de Itrio/uso terapéutico , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/farmacocinética , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Cintigrafía , Radiofármacos/farmacocinética , Especificidad por Sustrato , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Biother Radiopharm ; 19(4): 399-404, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15453954

RESUMEN

Arginine-glycine-aspartic acid (RGD) peptides preferentially bind to alphavbeta3 integrin, an integrin expressed on newly formed endothelial cells and on various tumor cells. When labeled with beta-emitting radionuclides, these peptides can be used for peptide-receptor radionuclide therapy of malignant tumors. These studies aimed to investigate whether tumor targeting and tumor therapy could be optimized by dose fractionation. The RGD-peptide DOTA-E-[c(RGDfK)]2 was labeled with 111In for biodistribution experiments and with 90Y for therapy experiments. In mice with NIH:OVCAR-3 ovarian carcinoma xenografts, optimal tumor uptake was obtained at peptide doses up to 1.0 microg (4.8 %ID/g). A peptide dose of 5 microg, required to administer the maximum tolerable dose (MTD) 90Y-DOTA-E-[c(RGDfK)]2, was administered as 5 portions of 1.0 microg. Tumor uptake of the fifth portion was significantly higher than that of the single 5.0 microg portion (3.3 %ID/g versus 2.1 %ID/g). The therapeutic efficacy of 37 MBq 90Y-DOTA-E-[c(RGDfK)]2 (1 x 5.0 microg) was compared with that of 37 MBq administered in five equal portions (5 x 1.0 microg). No difference in tumor growth between the fractionated and the nonfractionated therapy was observed. In conclusion, dose fractionation resulted in higher radiation doses. However, therapeutic efficacy of the radiolabeled peptide was not significantly improved by dose fractionation.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , Radioisótopos de Indio/uso terapéutico , Integrina alfaVbeta3/metabolismo , Oligopéptidos/metabolismo , Neoplasias Ováricas/radioterapia , Radiofármacos/metabolismo , Radioisótopos de Itrio/uso terapéutico , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Biomed Opt ; 18(10): 101319, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23933968

RESUMEN

Assays for blood levels of prostate-specific antigen (PSA), performed in prostate cancer detection, measure mostly inactive/complexed PSA and do not provide information regarding enzymatically active PSA, which is biologically more relevant. Thus, we designed and synthesized an enzymatically cleavable peptide sequence labeled with near-infrared (NIR) fluorophores (ex/em 740/770 nm) and coupled it to a pharmacokinetic modifier designed to improve its plasma kinetics. In its native state, the agent, PSA750 FAST™ (PSA750), is optically quenched (>95%) and only becomes fluorescent upon cleavage by active PSA, yielding a significant increase in signal. This activation is highly selective for PSA relative to a large panel of disease-relevant enzymes. Active PSA was detected in tumor frozen sections using PSA750 and this activity was abolished in the presence of the inhibitor, alpha-1 anti-chymotrypsin. In vivo imaging of tumor-bearing mice using fluorescence molecular tomography demonstrated a significantly higher fluorescent signal in PSA+ LNCaP tumors as compared to PSA- prostate cancer 3 tumors (13.0±3.7 versus 2.8±0.8 pmol, p=0.023). Ex vivo imaging of tumor sections confirms PSA750-derived NIR signal localization in nonvascular tissue. This is the first report that demonstrates the feasibility and effectiveness of noninvasive, real time, fluorescence molecular imaging of PSA enzymatic activity in prostate cancer.


Asunto(s)
Colorantes Fluorescentes/análisis , Imagen Molecular/métodos , Antígeno Prostático Específico/análisis , Tomografía Óptica/métodos , Análisis de Varianza , Animales , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Colorantes Fluorescentes/farmacocinética , Histocitoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Oligopéptidos/análisis , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/farmacocinética , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/química , Neoplasias de la Próstata/metabolismo
10.
PLoS One ; 7(11): e50860, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226406

RESUMEN

Carbonic anhydrase IX (CA IX) is a transmembrane protein that has been shown to be greatly upregulated under conditions of hypoxia in many tumor cell lines. Tumor hypoxia is associated with impaired efficacy of cancer therapies making CA IX a valuable target for preclinical and diagnostic imaging. We have developed a quantitative in vivo optical imaging method for detection of CA IX as a marker of tumor hypoxia based on a near-infrared (NIR) fluorescent derivative of the CA IX inhibitor acetazolamide (AZ). The agent (HS680) showed single digit nanomolar inhibition of CA IX as well as selectivity over other CA isoforms and demonstrated up to 25-fold upregulation of fluorescent CA IX signal in hypoxic versus normoxic cells, which could be blocked by 60%-70% with unlabeled AZ. CA IX negative cell lines (HCT-116 and MDA-MB-231), as well as a non-binding control agent on CA IX positive cells, showed low fluorescent signal under both conditions. In vivo FMT imaging showed tumor accumulation and excellent tumor definition from 6-24 hours. In vivo selectivity was confirmed by pretreatment of the mice with unlabeled AZ resulting in >65% signal inhibition. HS680 tumor signal was further upregulated >2X in tumors by maintaining tumor-bearing mice in a low oxygen (8%) atmosphere. Importantly, intravenously injected HS680 signal was co-localized specifically with both CA IX antibody and pimonidazole (Pimo), and was located away from non-hypoxic regions indicated by a Hoechst stain. Thus, we have established a spatial correlation of fluorescence signal obtained by non-invasive, tomographic imaging of HS680 with regions of hypoxia and CA IX expression. These results illustrate the potential of HS680 and combined with FMT imaging to non-invasively quantify CA IX expression as a hypoxia biomarker, crucial to the study of the underlying biology of hypoxic tumors and the development and monitoring of novel anti-cancer therapies.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Anhidrasas Carbónicas/metabolismo , Diagnóstico por Imagen/métodos , Neoplasias/enzimología , Neoplasias/patología , Imagen Óptica/métodos , Animales , Anhidrasa Carbónica IX , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Femenino , Fluorescencia , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Humanos , Ratones , Peso Molecular , Oxígeno/farmacología , Transporte de Proteínas/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Imaging ; 2012: 189254, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23119157

RESUMEN

Inflammation as a core pathological event of atherosclerotic lesions is associated with the secretion of cathepsin proteases and the expression of α(v)ß(3) integrin. We employed fluorescence molecular tomographic (FMT) noninvasive imaging of these molecular activities using cathepsin sensing (ProSense, CatB FAST) and α(v)ß(3) integrin (IntegriSense) near-infrared fluorescence (NIRF) agents. A statistically significant increase in the ProSense and IntegriSense signal was observed within the chest region of apoE(-/-) mice (P < 0.05) versus C57BL/6 mice starting 25 and 22 weeks on high cholesterol diet, respectively. In a treatment study using ezetimibe (7 mg/kg), there was a statistically significant reduction in the ProSense and CatB FAST chest signal of treated (P < 0.05) versus untreated apoE(-/-) mice at 31 and 21 weeks on high cholesterol diet, respectively. The signal of ProSense and CatB FAST correlated with macrophage counts and was found associated with inflammatory cells by fluorescence microscopy and flow cytometry of cells dissociated from aortas. This report demonstrates that cathepsin and α(v)ß(3) integrin NIRF agents can be used as molecular imaging biomarkers for longitudinal detection of atherosclerosis, and cathepsin agents can monitor anti-inflammatory effects of ezetimibe with applications in preclinical testing of therapeutics and potentially for early diagnosis of atherosclerosis in patients.

12.
Int J Mol Imaging ; 2011: 581406, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21941648

RESUMEN

We developed a neutrophil elastase-specific near-infrared fluorescence imaging agent, which, combined with fluorescence molecular tomographic imaging, allowed us to detect and quantify neutrophil elastase activity in vivo, in real time, and noninvasively in an acute model of lung injury (ALI). Significantly higher fluorescent signal was quantified in mice with LPS/fMLP-induced ALI as compared to healthy controls, correlating with increases in the number of bronchoalveolar lavage cells, neutrophils, and elastase activity. The agent was significantly activated ex vivo in lung sections from ALI but not from control mice, and this activation was ablated by the specific inhibitor sivelestat. Treatment with the specific inhibitor sivelestat significantly reduced lung signal in mice with ALI. These results underscore the unique ability of fluorescence molecular imaging to quantify specific molecular processes in vivo, crucial for understanding the mechanisms underlying disease progression and for assessing and monitoring novel pharmacological interventions.

13.
Mol Imaging Biol ; 12(5): 488-99, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19960268

RESUMEN

PURPOSE: Integrins, especially α(v)ß(3) and α(v)ß(5), are upregulated in tumor cells and activated endothelial cells and as such, serve as cancer biomarkers. We developed a novel near-infrared-labeled optical agent for the in vivo detection and quantification of α(v)ß(3)/α(v)ß(5). PROCEDURES: A small peptidomimetic α(v)ß(3) antagonist was synthesized, coupled to a near-infrared fluorescent (NIRF) dye, and tested for binding specificity using integrin-overexpressing cells, inhibition of vitronectin-mediated cell attachment, binding to tumor and endothelial cells in vitro, and competition studies. Pharmacokinetics, biodistribution, specificity of tumor targeting, and the effect of an antiangiogenic treatment were assessed in vivo. RESULTS: The integrin NIRF agent showed strong selectivity towards α(v)ß(3/)α(v)ß(5) in vitro and predominant tumor distribution in vivo, allowing noninvasive and real-time quantification of integrin signal in tumors. Antiangiogenic treatment significantly inhibited integrin signal in vivo but had no effect on a cathepsin-cleavable NIR agent. Simultaneous imaging revealed different patterns of distribution reflecting the underlying differences in integrin and cathepsin biology during tumor progression. CONCLUSIONS: NIRF-labeled integrin antagonists allow noninvasive molecular fluorescent imaging and quantification of tumors in vivo, improving and providing more refined approaches for cancer detection and treatment monitoring.


Asunto(s)
Antineoplásicos/farmacocinética , Integrinas/metabolismo , Péptido Hidrolasas/metabolismo , Tomografía/métodos , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Fluorescencia , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Distribución Tisular , Trasplante Heterólogo
14.
Bioconjug Chem ; 18(4): 1266-79, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17579472

RESUMEN

The integrin receptor alphavbeta3 is overexpressed on the endothelial cells of growing tumors and on some tumor cells themselves. A radiolabeled alphavbeta3 antagonists belonging to the quinolin-4-one class of peptidomimetics (TA138) was previously shown to exhibit high affinity for integrin alphavbeta3 and high selectivity versus other integrin receptors. 111In-TA138 exhibited high tumor uptake in the c-neu Oncomouse mammary adenocarcinoma model and produced excellent scintigraphic images. This study describes the synthesis of eight divalent versions of TA138 and their evaluation as potential tumor radiotherapeutic agents. The two main variables in this study were the length of the spacer bridging the biotargeting moieties and the total negative charge of the molecules imparted by the cysteic acid pharmacokinetic modifiers. Receptor affinity was evaluated in a panel of integrin receptor affinity assays, and biodistribution studies using the 111In-labeled derivatives were carried out in the c-neu Oncomouse model. All divalent agents maintained the high receptor affinity and selectivity of TA138, and six of the eight 111In derivatives exhibited blood clearance that was faster than 111In-TA138 at 24 h postinjection (PI). All divalent agents exhibited tumor uptake and retention at 24 h PI that was higher than 111In-TA138. Tumor/organ ratios were improved for most of the divalent agents at 24 h PI in critical nontarget organs marrow, kidney, and liver, with the agents having intermediate-length spacers (29-43 A) showing the largest improvement. As an example, 111In-15 showed tumor uptake of 14.3% ID/g at 24 h PI and tumor/organ ratios as follows: marrow, 3.24; kidney, 7.29; liver, 8.51. A comparison of therapeutic indices for 90Y-TA138 and 177Lu-15 indicate an improved therapeutic index for the divalent agent. The implications for radiotherapeutic applications and the mechanism of this multivalent effect are discussed.


Asunto(s)
Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Radioisótopos de Indio/química , Integrina alfaVbeta3/antagonistas & inhibidores , Lutecio/farmacocinética , Radioisótopos/farmacocinética , Radiofármacos/farmacocinética , Sulfonamidas/farmacocinética , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/metabolismo , Adenocarcinoma/radioterapia , Animales , Unión Competitiva , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Diagnóstico por Imagen , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , Radioisótopos de Indio/farmacocinética , Radioisótopos de Indio/uso terapéutico , Integrina alfaVbeta3/metabolismo , Lutecio/química , Lutecio/uso terapéutico , Ratones , Péptidos/química , Péptidos/farmacocinética , Péptidos/uso terapéutico , Radioisótopos/química , Radioisótopos/uso terapéutico , Cintigrafía , Radiofármacos/química , Radiofármacos/uso terapéutico , Sulfonamidas/química , Sulfonamidas/uso terapéutico , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA