Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nanomedicine (Lond) ; 17(22): 1649-1662, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36547231

RESUMEN

Aim: To characterize the pharmacokinetics of deferoxamine-conjugated nanoparticles (DFO-NPs), a novel nanochelator for removing excess iron. Materials & methods: The pharmacokinetics of DFO-NPs were evaluated in Sprague-Dawley rats at three doses (3.3, 10 and 30 µmol/kg) after intravenous and subcutaneous administration. Results: DFO-NPs exhibited a biphasic concentration-time profile after intravenous administration with a short terminal half-life (2.0-3.2 h), dose-dependent clearance (0.111-0.179 l/h/kg), minimal tissue distribution and exclusive renal excretion with a possible saturable reabsorption mechanism. DFO-NPs after subcutaneous administration exhibited absorption-rate-limited kinetics with a prolonged half-life (5.7-10.1 h) and favorable bioavailability (47-107%). Conclusion: DFO-NPs exhibit nonlinear pharmacokinetics with increasing dose, and subcutaneous administration substantially improves drug exposure, thereby making it a clinically viable administration route for iron chelation.


Iron is an essential metal nutrient, but excess iron produces toxic effects that damage multiple organs including the heart, liver and pancreas. Deferoxamine (DFO) is a US FDA-approved drug for treating iron overload, but its use is limited by serious adverse effects and an inconvenient daily dose scheme. The recent development of a DFO-based nanomedicine (DFO-NP) has shown promise in treating iron overload in animals and was safer in animals. Before this new drug can be given to humans, how it is absorbed into the body, processed in the body and removed from the body when given in different amounts and dose routes must be determined. In this study, we tested the absorption, distribution and removal of DFO-NPs after intravenous and subcutaneous injection in rats. This study showed that DFO-NPs behave differently when changing the dose and that subcutaneous injection makes the drug stay in the body longer without ill effect, which means it could be given to patients this way.


Asunto(s)
Deferoxamina , Sobrecarga de Hierro , Ratas , Animales , Deferoxamina/farmacocinética , Deferoxamina/uso terapéutico , Quelantes del Hierro/farmacocinética , Quelantes del Hierro/uso terapéutico , Distribución Tisular , Ratas Sprague-Dawley , Sobrecarga de Hierro/tratamiento farmacológico
2.
Mol Pharm ; 18(2): 593-609, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32926630

RESUMEN

Deferoxamine mesylate (DFO) is an FDA-approved, hexadentate iron chelator routinely used to alleviate systemic iron burden in thalassemia major and sickle cell patients. Iron accumulation in these disease states results from the repeated blood transfusions required to manage these conditions. Iron accumulation has also been implicated in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), and secondary injury following intracerebral hemorrhage (ICH). Chelation of brain iron is thus a promising therapeutic strategy for improving behavioral outcomes and slowing neurodegeneration in the aforementioned disease states, though the effectiveness of DFO treatment is limited on several accounts. Systemically administered DFO results in nonspecific toxicity at high doses, and the drug's short half-life leads to low patient compliance. Mixed reports of DFO's ability to cross the blood-brain barrier (BBB) also appear in literature. These limitations necessitate novel DFO formulations prior to the drug's widespread use in managing neurodegeneration. Herein, we discuss the various dosing regimens and formulations employed in intranasal (IN) or systemic DFO treatment, as well as the physiological and behavioral outcomes observed in animal models of AD, PD, and ICH. The clinical progress of chelation therapy with DFO in managing neurodegeneration is also evaluated. Finally, the elimination of intranasally administered particles via the glymphatic system and efflux transporters is discussed. Abundant preclinical evidence suggests that intranasal DFO treatment improves memory retention and behavioral outcome in rodent models of AD, PD, and ICH. Several other biochemical and physiological metrics, such as tau phosphorylation, the survival of tyrosine hydroxylase-positive neurons, and infarct volume, are also positively affected by intranasal DFO treatment. However, dosing regimens are inconsistent across studies, and little is known about brain DFO concentration following treatment. Systemic DFO treatment yields similar results, and some complex formulations have been developed to improve permeability across the BBB. However, despite the success in preclinical models, clinical translation is limited with most clinical evidence investigating DFO treatment in ICH patients, where high-dose treatment has proven dangerous and dosing regimens are not consistent across studies. DFO is a strong drug candidate for managing neurodegeneration in the aging population, but before it can be routinely implemented as a therapeutic agent, dosing regimens must be standardized, and brain DFO content following drug administration must be understood and controlled via novel formulations.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Deferoxamina/administración & dosificación , Portadores de Fármacos/química , Enfermedad de Parkinson/tratamiento farmacológico , Sideróforos/administración & dosificación , Administración Intranasal , Enfermedad de Alzheimer/patología , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/patología , Deferoxamina/farmacocinética , Modelos Animales de Enfermedad , Semivida , Humanos , Inyecciones Intramusculares , Inyecciones Intraventriculares , Inyecciones Espinales , Inyecciones Subcutáneas , Hierro/metabolismo , Cumplimiento de la Medicación , Nanopartículas/química , Mucosa Nasal/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/patología , Permeabilidad , Sideróforos/farmacocinética , Distribución Tisular
3.
ACS Appl Mater Interfaces ; 12(51): 56792-56804, 2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33306342

RESUMEN

Radionuclide-functionalized drug delivery vehicles capable of being imaged via positron emission tomography (PET) are of increasing interest in the biomedical field as they can reveal the in vivo behavior of encapsulated therapeutics with high sensitivity. However, the majority of current PET-guided theranostic agents suffer from poor retention of radiometal over time, low drug loading capacities, and time-limited PET imaging capability. To overcome these challenges, we have developed hollow microcapsules with a thin (<100 nm) multilayer shell as advanced theranostic delivery systems for multiday PET tracking in vivo. The 3 µm capsules were fabricated via the aqueous multilayer assembly of a natural antioxidant, tannic acid (TA), and a poly(N-vinylpyrrolidone) (PVPON) copolymer containing monomer units functionalized with deferoxamine (DFO) to chelate the 89Zr radionuclide, which has a half-life of 3.3 days. We have found using radiochromatography that (TA/PVPON-DFO)6 capsules retained on average 17% more 89Zr than their (TA/PVPON)6 counterparts, which suggests that the covalent attachment of the DFO to PVPON provides stable 89Zr chelation. In vivo PET imaging studies performed in mice demonstrated that excellent stability and imaging contrast were still present 7 days postinjection. Animal biodistribution analyses showed that capsules primarily accumulated in the spleen, liver, and lungs with negligible accumulation in the femur, with the latter confirming the stable binding of the radiotracer to the capsule walls. The application of therapeutic ultrasound (US) (60 s of 20 kHz US at 120 W cm-2) to Zr-functionalized capsules could release the hydrophilic anticancer drug doxorubicin from the capsules in the therapeutic amounts. Polymeric capsules with the capability of extended in vivo PET-based tracking and US-induced drug release provide an advanced platform for development of precision-targeted therapeutic carriers and could aid in the development of more effective drug delivery systems.


Asunto(s)
Antineoplásicos/uso terapéutico , Quelantes/química , Medios de Contraste/química , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Resinas Acrílicas/química , Resinas Acrílicas/farmacocinética , Animales , Cápsulas , Quelantes/farmacocinética , Medios de Contraste/farmacocinética , Deferoxamina/química , Deferoxamina/farmacocinética , Portadores de Fármacos/farmacocinética , Femenino , Ratones Endogámicos BALB C , Tomografía de Emisión de Positrones/métodos , Povidona/química , Povidona/farmacocinética , Medicina de Precisión/métodos , Radioisótopos/química , Taninos/química , Taninos/farmacocinética , Ondas Ultrasónicas , Circonio/química
4.
Int J Mol Sci ; 21(14)2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32679799

RESUMEN

Proteins, as a major component of organisms, are considered the preferred biomaterials for drug delivery vehicles. Hemoglobin (Hb) has been recently rediscovered as a potential drug carrier, but its use for biomedical applications still lacks extensive investigation. To further explore the possibility of utilizing Hb as a potential tumor targeting drug carrier, we examined and compared the biodistribution of Hb in healthy and lung tumor-bearing mice, using for the first time 89Zr labelled Hb in a positron emission tomography (PET) measurement. Hb displays a very high conjugation yield in its fast and selective reaction with the maleimide-deferoxamine (DFO) bifunctional chelator. The high-resolution X-ray structure of the Hb-DFO complex demonstrated that cysteine ß93 is the sole attachment moiety to the αß-protomer of Hb. The Hb-DFO complex shows quantitative uptake of 89Zr in solution as determined by radiochromatography. Injection of 0.03 mg of Hb-DFO-89Zr complex in healthy mice indicates very high radioactivity in liver, followed by spleen and lungs, whereas a threefold increased dosage results in intensification of PET signal in kidneys and decreased signal in liver and spleen. No difference in biodistribution pattern is observed between naïve and tumor-bearing mice. Interestingly, the liver Hb uptake did not decrease upon clodronate-mediated macrophage depletion, indicating that other immune cells contribute to Hb clearance. This finding is of particular interest for rapidly developing clinical immunology and projects aiming to target, label or specifically deliver agents to immune cells.


Asunto(s)
Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos , Hemoglobinas/farmacocinética , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Animales , Línea Celular Tumoral , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Deferoxamina/análogos & derivados , Deferoxamina/farmacocinética , Portadores de Fármacos/química , Femenino , Hemoglobinas/química , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radioisótopos/química , Radioisótopos/farmacocinética , Distribución Tisular , Circonio/química , Circonio/farmacocinética
5.
Nat Commun ; 10(1): 5134, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31723130

RESUMEN

Iron chelators have been widely used to remove excess toxic iron from patients with secondary iron overload. However, small molecule-based iron chelators can cause adverse side effects such as infection, gastrointestinal bleeding, kidney failure, and liver fibrosis. Here we report renal clearable nanochelators for iron overload disorders. First, after a singledose intravenous injection, the nanochelator shows favorable pharmacokinetic properties, such as kidney-specific biodistribution and rapid renal excretion (>80% injected dose in 4 h), compared to native deferoxamine (DFO). Second, subcutaneous (SC) administration of nanochelators improves pharmacodynamics, as evidenced by a 7-fold increase in efficiency of urinary iron excretion compared to intravenous injection. Third, daily SC injections of the nanochelator for 5 days to iron overload mice and rats decrease iron levels in serum and liver. Furthermore, the nanochelator significantly reduces kidney damage caused by iron overload without demonstrating DFO's own nephrotoxicity. This renal clearable nanochelator provides enhanced efficacy and safety.


Asunto(s)
Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Riñón/patología , Nanopartículas/química , Animales , Deferoxamina/farmacocinética , Deferoxamina/uso terapéutico , Deferoxamina/toxicidad , Quelantes del Hierro/química , Quelantes del Hierro/farmacología , Masculino , Ratones , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Distribución Tisular
6.
Eur J Nucl Med Mol Imaging ; 46(9): 1966-1977, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31161258

RESUMEN

PURPOSE: Currently, the most commonly used chelator for labelling antibodies with 89Zr for immunoPET is desferrioxamine B (DFO). However, preclinical studies have shown that the limited in vivo stability of the 89Zr-DFO complex results in release of 89Zr, which accumulates in mineral bone. Here we report a novel chelator DFOcyclo*, a preorganized extended DFO derivative that enables octacoordination of the 89Zr radiometal. The aim was to compare the in vitro and in vivo stability of [89Zr]Zr-DFOcyclo*, [89Zr]Zr-DFO* and [89Zr]Zr-DFO. METHODS: The stability of 89Zr-labelled chelators alone and after conjugation to trastuzumab was evaluated in human plasma and PBS, and in the presence of excess EDTA or DFO. The immunoreactive fraction, IC50, and internalization rate of the conjugates were evaluated using HER2-expressing SKOV-3 cells. The in vivo distribution was investigated in mice with subcutaneous HER2+ SKOV-3 or HER2- MDA-MB-231 xenografts by PET/CT imaging and quantitative ex vivo tissue analyses 7 days after injection. RESULTS: 89Zr-labelled DFO, DFO* and DFOcyclo* were stable in human plasma for up to 7 days. In competition with EDTA, DFO* and DFOcyclo* showed higher stability than DFO. In competition with excess DFO, DFOcyclo*-trastuzumab was significantly more stable than the corresponding DFO and DFO* conjugates (p < 0.001). Cell binding and internalization were similar for the three conjugates. In in vivo studies, HER2+ SKOV-3 tumour-bearing mice showed significantly lower bone uptake (p < 0.001) 168 h after injection with [89Zr]Zr-DFOcyclo*-trastuzumab (femur 1.5 ± 0.3%ID/g, knee 2.1 ± 0.4%ID/g) or [89Zr]Zr-DFO*-trastuzumab (femur 2.0 ± 0.3%ID/g, knee 2.68 ± 0.4%ID/g) than after injection with [89Zr]Zr-DFO-trastuzumab (femur 4.5 ± 0.6%ID/g, knee 7.8 ± 0.6%ID/g). Blood levels, tumour uptake and uptake in other organs were not significantly different at 168 h after injection. HER2- MDA-MB-231 tumour-bearing mice showed significantly lower tumour uptake (p < 0.001) after injection with [89Zr]Zr-DFOcyclo*-trastuzumab (16.2 ± 10.1%ID/g) and [89Zr]Zr-DFO-trastuzumab (19.6 ± 3.2%ID/g) than HER2+ SKOV-3 tumour-bearing mice (72.1 ± 14.6%ID/g and 93.1 ± 20.9%ID/g, respectively), while bone uptake was similar. CONCLUSION: 89Zr-labelled DFOcyclo* and DFOcyclo*-trastuzumab showed higher in vitro and in vivo stability than the current commonly used 89Zr-DFO-trastuzumab. DFOcyclo* is a promising candidate to become the new clinically used standard chelator for 89Zr immunoPET.


Asunto(s)
Deferoxamina/química , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radioisótopos/química , Circonio/química , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Deferoxamina/farmacocinética , Femenino , Humanos , Ratones , Distribución Tisular
7.
J Control Release ; 281: 139-177, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29772289

RESUMEN

According to the Alzheimer Association Report (2017), Alzheimer's disease (AD) is the 6th primary cause of death in the USA, which affects nearly 5.5 million people. In the year 2017 itself, the cost of AD treatment in the USA has been reported to rise to $259 billion. This statistic shows the severity of the disease in the USA which is very much similar across the globe. On the other hand, the treatment remains limited to a few conventional oral medications (approved by FDA). These are mainly acting superficially from mild to the moderate AD. The therapeutic efficacy of the drug is not only affected by its reduced concentration in the brain owing to the existence of blood-brain-barrier (BBB) but also due to its low brain permeability. In this context, the intranasal (IN) route of drug administration has emerged as an alternative route over the systemic (oral and parenteral) drug delivery to the brain. The delivery of the drug via an IN route offers various advantages over systemic drug delivery system, as it directly delivers the drug into the brain via olfactory route. Presence of drug in the olfactory bulb, in turn, increases the drug bioavailability in the brain and reduces the drug degradation as well as wastage of the drug through` systemic clearance. However, there is also some limitation associated with IN like poor drug permeation through the nasal mucosa and mucociliary clearance. The delivery system various through novel strategies (nano drug carrier system, colloidal carriers, mucoadhesive devices, controlled delivery system, pro-drug, etc.) are adapted to overcome the above-stated limitations. Although, after all, such successful research claims, very few of the nose-to-brain drug delivery of anti-AD drugs have gained market approval due to lack of sufficient clinical evidence. Onzetra Xsail® is one such marketed preparations approved for IN delivery used for the treatment of a brain disorder; migraine. In the field of patents also, no work is found which could present sufficient experimental findings to support its clinical safety profile. It also underlines the fact that majority of work related to the nose-to-brain delivery of anti-AD drugs is limited only up to preclinical studies. In this review article, we have discussed the latest works on various novel formulations loaded with various anti-Alzheimer agents. These agents include galantamine, deferoxamine, tacrine, tarenflurbil, rivastigmine, risperidone, curcumin, quercetin, piperine, insulin, etc. and various peptides towards the development of a promising IN drug delivery system for the treatment of AD. Through this review article, we want to drag the attention of the researchers working in this field towards the challenges and hurdles of practical applicability IN delivery of anti-AD drugs. Moreover, the attention towards the clinical studies will ease the approval process for the administration of anti-Alzheimer drugs via IN route.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Barrera Hematoencefálica/metabolismo , Encéfalo , Portadores de Fármacos/farmacocinética , Nanopartículas/química , Nariz , Administración Intranasal , Animales , Disponibilidad Biológica , Deferoxamina/administración & dosificación , Deferoxamina/farmacocinética , Deferoxamina/uso terapéutico , Donepezilo/administración & dosificación , Donepezilo/farmacocinética , Donepezilo/uso terapéutico , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/uso terapéutico , Liberación de Fármacos , Galantamina/administración & dosificación , Galantamina/farmacocinética , Galantamina/uso terapéutico , Humanos , Depuración Mucociliar , Mucosa Nasal/metabolismo , Bulbo Olfatorio/metabolismo , Risperidona/administración & dosificación , Risperidona/farmacocinética , Risperidona/uso terapéutico , Distribución Tisular
8.
ACS Nano ; 12(5): 4123-4139, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29617109

RESUMEN

Excess iron deposition in the brain often causes oxidative stress-related damage and necrosis of dopaminergic neurons in the substantia nigra and has been reported to be one of the major vulnerability factors in Parkinson's disease (PD). Iron chelation therapy using deferoxamine (DFO) may inhibit this nigrostriatal degeneration and prevent the progress of PD. However, DFO shows very short half-life in vivo and hardly penetrates the blood brain barrier (BBB). Hence, it is of great interest to develop DFO formulations for safe and efficient intracerebral drug delivery. Herein, we report a polymeric nanoparticle system modified with brain-targeting peptide rabies virus glycoprotein (RVG) 29 that can intracerebrally deliver DFO. The nanoparticle system penetrates the BBB possibly through specific receptor-mediated endocytosis triggered by the RVG29 peptide. Administration of these nanoparticles significantly decreased iron content and oxidative stress levels in the substantia nigra and striatum of PD mice and effectively reduced their dopaminergic neuron damage and as reversed their neurobehavioral deficits, without causing any overt adverse effects in the brain or other organs. This DFO-based nanoformulation holds great promise for delivery of DFO into the brain and for realizing iron chelation therapy in PD treatment.


Asunto(s)
Encéfalo/metabolismo , Deferoxamina/administración & dosificación , Sistemas de Liberación de Medicamentos , Glicoproteínas/química , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Fragmentos de Péptidos/química , Proteínas Virales/química , Animales , Encéfalo/efectos de los fármacos , Deferoxamina/farmacocinética , Deferoxamina/farmacología , Deferoxamina/uso terapéutico , Glicoproteínas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/metabolismo , Fragmentos de Péptidos/administración & dosificación , Sideróforos/administración & dosificación , Sideróforos/farmacocinética , Sideróforos/farmacología , Sideróforos/uso terapéutico , Proteínas Virales/administración & dosificación
9.
Int J Pharm ; 538(1-2): 79-86, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29341909

RESUMEN

Deferoxamine (DFO) to treat iron overload (IO) has been limited by toxicity issues and short circulation times and it would be desirable to prolong circulation to improve non-transferrin bound iron (NTBI) chelation. In addition, DFO is currently unable to efficiently target the large pool of iron in the liver and spleen. Nanogel-Deferoxamine conjugates (NG-DFO) can prove useful as a model to investigate the pharmacokinetic (PK) properties and biodistribution (BD) behavior of iron-chelating macromolecules and their overall effect on serum ferritin levels. NG-DFO reduced the cytotoxicity of DFO and significantly reduced cellular ferritin levels in IO macrophages in vitro. PK/BD studies in normal rats revealed that NG-DFO displayed prolonged circulation and preferential accumulation into the liver and spleen. IO mice treated with NG1-DFO presented significantly lower levels of serum ferritin compared to DFO. Total renal and fecal elimination data point to the need to balance prolonged circulation with controlled degradation to accelerate clearance of iron-chelating macromolecules.


Asunto(s)
Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Modelos Biológicos , Animales , Deferoxamina/farmacocinética , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Femenino , Ferritinas/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Quelantes del Hierro/farmacocinética , Quelantes del Hierro/farmacología , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Ratas , Bazo/metabolismo , Distribución Tisular
10.
Nucl Med Biol ; 58: 1-7, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29291493

RESUMEN

INTRODUCTION: Zirconium-89 (89Zr, t1/2=78.4h) liquid target (LT) production offers an approach to introduce this positron-emitting isotope to cyclotron centres without the need for a separate solid target (ST) production set up. We compared the production, purification, and antibody radiolabeling yields of 89Zr-(LT) and 89Zr-(ST), and assessed the feasibility of 89Zr-(LT) for preclinical PET/CT. METHODS: 89Zr-(ST) production was performed with an 89Y foil on a TR 19 cyclotron at 13.8MeV. For LT production; an aqueous solution of yttrium nitrate (Y(NO3)3·6H2O) was irradiated on a TR 13 cyclotron at 12MeV. 89Zr was purified from the ST or LT material with hydroxamate resin, and used to radiolabel p-SCN-Bn-Deferoxamine (DFO)-conjugated Trastuzumab. MicroPET-CT imaging was performed at 1, 3 and 5days post-injection of 89Zr-DFO-Trastuzumab from ST or LT with biodistribution analysis on day 5. RESULTS: Irradiation of the ST yielded 2.88±1.07GBq/µA with a beam current of 14.0±3.8µA and irradiation time of 137±48min at end of bombardment while LT yielded 0.27±0.05GBq/µA with a beam current of 9.9±2.2µA and irradiation time of 221±29min. Radiolabeling of DFO-Trastuzumab with 89Zr-(ST) or 89Zr-(LT) was successful with purity>97% and specific activity>0.12MBq/µg (of antibody). MicroPET-CT imaging and biodistribution profiles showed similar uptake of 89Zr-(ST)-DFO-Trastuzumab and 89Zr-(LT)-DFO-Trastuzumab in tumor and all organs of interest. CONCLUSION: 89Zr-(LT) was effectively used to prepare antibody bioconjugates with specific activities suitable for small animal imaging. PET imaging and biodistribution revealed similar behaviours between bioconjugates labeled with 89Zr produced from the two target systems. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: These results have important implications for the production of PET isotopes such as 89Zr to cyclotron facilities with only LT capabilities - such as most clinical centres - expanding the availability of 89Zr-immunoPET.


Asunto(s)
Deferoxamina/química , Marcaje Isotópico/métodos , Radioisótopos/química , Trastuzumab/química , Circonio/química , Animales , Deferoxamina/farmacocinética , Femenino , Ratones , Tomografía Computarizada por Tomografía de Emisión de Positrones , Distribución Tisular , Trastuzumab/farmacocinética
11.
Colloids Surf B Biointerfaces ; 161: 129-138, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29055865

RESUMEN

In this study, an injectable chitosan-hyaluronic acid (CS-HA) based hydrogel was designed incorporating pro-angiogenic molecule, deferoxamine loaded PLGA nanoparticles (DFO NPs), for enhancing angiogenesis. DFO-NPs were prepared by double emulsion solvent diffusion technique and characterized for their physicochemical properties. The DLS and SEM analysis showed an average particle size of 220±71nm with spherical morphology and the encapsulation efficiency was found to be 30±5%. An ECM mimicking chitosan-hyaluronic acid (CS-HA) coacervate hydrogel was prepared. Both free DFO and DFO NPs were entrapped into the prepared CS-HA composite hydrogel. The hydrogels were characterized by SEM, FTIR and Rheology. Addition of DFO NPs did not affect the injectablility and flowability of developed hydrogels. In vitro DFO release from the prepared composite hydrogels showed controlled release over a period of 10days. Both the hydrogel systems showed excellent cyto-compatability and good cell proliferation for rASCs as well as HUVECs. The DFO and DFO NPs loaded composite hydrogels revealed effective tube formation in comparison with control hydrogels without DFO and DFO NPs. The in vivo angiogenic evaluation of the free DFO and DFO NPs (0.025%w/w) loaded composite hydrogels were studied by injecting the developed hydrogel subcutaneously into mice for 2-4 weeks. The DFO NPs loaded composite hydrogel had enhanced neovascularization when compared to control gels. Thus, the developed DFO NPs loaded composite hydrogel could potentially be used for therapeutic angiogenesis.


Asunto(s)
Quitosano/química , Deferoxamina/administración & dosificación , Ácido Hialurónico/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas/administración & dosificación , Neovascularización Fisiológica/efectos de los fármacos , Animales , Células Cultivadas , Deferoxamina/química , Deferoxamina/farmacocinética , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Liberación de Fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Inyecciones , Ácido Láctico/química , Ratones Endogámicos BALB C , Nanopartículas/química , Tamaño de la Partícula , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
12.
Mol Imaging ; 16: 1536012117737010, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29098927

RESUMEN

Radiotracers incorporating the urea-based Glu-NH-C(O)-NH-Lys group have gained prominence due to their role in targeting prostate-specific membrane antigen (PSMA)-a clinical biomarker of prostate cancer. Here, the synthesis, radiolabeling, and in vitro and in vivo characterization of two 68Ga-radiolabeled Glu-NH-C(O)-NH-Lys radiotracers conjugated to the desferrioxamine B (DFO) chelate were evaluated. Two linker groups based on amide bond and thiourea coupling chemistries were employed to develop 68Ga-DFO-Nsucc-PSMA (68Ga-4) and 68Ga-DFO- pNCS-Bn-PSMA (68Ga-7), respectively. Radiosynthesis proceeded quantitatively at room temperature with high radiochemical yields, chemical/radiochemical purities, and specific activities. Pharmacokinetic profiles of 68Ga-4 and 68Ga-7 were assessed using positron-emission tomography (PET) in mice bearing subcutaneous LNCaP tumors. Data were compared to the current clinical benchmark radiotracer 68Ga-HBED-CC-PSMA (68Ga-1) (HBED = N,N'-Bis(2-hydroxy-5-(ethylene-beta-carboxy)benzyl)ethylenediamine N,N'-diacetic acid). Results indicated that the target binding affinity, protein association, blood pool and background organ clearance properties, and uptake in PSMA-positive lesions are strongly dependent on the nature of the chelate, the linker, and the spacer groups. Protein dissociation constants ( Kd values) were found to be predictive of pharmacokinetics in vivo. Compared to 68Ga-1, 68Ga-4 and 68Ga-7 resulted in decreased tumor uptake but enhanced blood pool clearance and reduced residence time in the kidney. The study highlights the importance of maximizing protein binding affinity during radiotracer optimization.


Asunto(s)
Deferoxamina/química , Radioisótopos de Galio/química , Tomografía de Emisión de Positrones , Antígeno Prostático Específico/metabolismo , Radiofármacos/química , Animales , Proteínas Sanguíneas/metabolismo , Línea Celular Tumoral , Deferoxamina/síntesis química , Deferoxamina/farmacocinética , Ácido Edético/análogos & derivados , Ácido Edético/química , Radioisótopos de Galio/farmacocinética , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Unión Proteica , Radioquímica , Radiofármacos/síntesis química , Radiofármacos/farmacocinética
13.
J Control Release ; 264: 160-168, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-28843831

RESUMEN

Overexpression of tissue factor (TF) has been associated with increased tumor growth, tumor angiogenesis, and metastatic potential in many malignancies, including pancreatic cancer. Additionally, high TF expression was shown to strongly correlate with poor prognoses and decreased survival in pancreatic cancer patients. Herein, we exploited the potential targeting of TF for positron emission tomography (PET) imaging of pancreatic cancer. The TF-targeted tracer was developed through radiolabeling of the anti-human TF monoclonal antibody (ALT-836) with 89Zr. The tracer was characterized by fluorescence microscopy and flow cytometry assays in BXPC-3 and PANC-1 cells, two pancreatic cancer cell lines with high and low TF expression levels, respectively. Non-invasive PET scans were acquired in tumor-bearing mice injected with 89Zr-Df-ALT-836. Additionally, ex vivo biodistribution, blocking, and histological studies were performed to establish the affinity and specificity of 89Zr-Df-ALT-836 for TF in vivo. 89Zr-labeling of Df-ALT-836 was achieved in high yield and good specific activity. Flow cytometry and microscopy studies revealed no detectable difference in TF-binding affinity between ALT-836 and Df-ALT-836 in vitro. Longitudinal PET scans unveiled a lasting and prominent 89Zr-Df-ALT-836 uptake in BXPC-3 tumors (peak at 31.5±6.0%ID/g at 48h post-injection; n=3), which was significantly abrogated (2.3±0.5%ID/g at 48h post-injection; n=3) when mice were pre-injected with a blocking dose (50mg/kg) of unlabeled ALT-836. Ex vivo biodistribution data confirmed the accuracy of the PET results, and histological analysis correlated high tumor uptake with in situ TF expression. Taken together, these results attest to the excellent affinity and TF-specificity of 89Zr-Df-ALT-836. With elevated, persistent, and specific accumulation in TF-positive BXPC-3 tumors, PET imaging using 89Zr-Df-ALT-836 promises to open new avenues for improving future diagnosis, stratification, and treatment response assessment in pancreatic cancer patients.


Asunto(s)
Deferoxamina , Inmunoglobulina G , Neoplasias Pancreáticas/metabolismo , Radioisótopos , Radiofármacos , Proteínas Recombinantes , Tromboplastina/metabolismo , Circonio , Animales , Línea Celular Tumoral , Deferoxamina/administración & dosificación , Deferoxamina/química , Deferoxamina/farmacocinética , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/química , Riñón/metabolismo , Hígado/metabolismo , Ratones Desnudos , Tomografía de Emisión de Positrones/métodos , Radioisótopos/administración & dosificación , Radioisótopos/química , Radioisótopos/farmacocinética , Radiofármacos/administración & dosificación , Radiofármacos/química , Radiofármacos/farmacocinética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Sideróforos/administración & dosificación , Sideróforos/química , Sideróforos/farmacocinética , Bazo/metabolismo , Distribución Tisular , Circonio/administración & dosificación , Circonio/química , Circonio/farmacocinética
14.
Bioconjug Chem ; 28(9): 2211-2223, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28767228

RESUMEN

Immuno-positron emission tomography (immunoPET) with 89Zr-labeled antibodies has shown great potential in cancer imaging. It can provide important information about the pharmacokinetics and tumor-targeting properties of monoclonal antibodies and may help in anticipating on toxicity. Furthermore, it allows accurate dose planning for individualized radioimmunotherapy and may aid in patient selection and early-response monitoring for targeted therapies. The most commonly used chelator for 89Zr is desferrioxamine (DFO). Preclinical studies have shown that DFO is not an ideal chelator because the 89Zr-DFO complex is partly unstable in vivo, which results in the release of 89Zr from the chelator and the subsequent accumulation of 89Zr in bone. This bone accumulation interferes with accurate interpretation and quantification of bone uptake on PET images. Therefore, there is a need for novel chelators that allow more stable complexation of 89Zr. In this Review, we will describe the most recent developments in 89Zr radiochemistry, including novel chelators and site-specific conjugation methods.


Asunto(s)
Quelantes/química , Inmunoconjugados/química , Neoplasias/diagnóstico , Tomografía de Emisión de Positrones/métodos , Circonio/química , Animales , Quelantes/farmacocinética , Deferoxamina/química , Deferoxamina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Humanos , Inmunoconjugados/farmacocinética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Radioquímica/métodos , Radioisótopos/química , Radioisótopos/farmacocinética , Circonio/farmacocinética
15.
Nucl Med Biol ; 52: 32-41, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28602965

RESUMEN

INTRODUCTION: Deferoxamine (DFO) is a siderophore that bacteria use to scavenge iron and could serve as a targeting vector to image bacterial infection where current techniques have critical limitations. [67Ga]-DFO, which is a mimetic of the corresponding iron complex, is taken up by bacteria in culture, however in vivo it clears too rapidly to allow for imaging of infection. In response, we developed several new DFO derivatives to identify those that accumulate in bacteria, and at sites of infection, and that could potentially have improved pharmacokinetics. METHODS: A library of DFO derivatives was synthesized by functionalizing the terminal amine group of DFO using three different carbamate-forming reactions. Uptake of [67Ga]-DFO and the 67Ga-labeled derivatives by bacteria and the biodistribution of lead compounds were studied. RESULTS: 67Ga-labeled DFO derivatives were prepared and isolated in >90% radiochemical yield and >95% radiochemical purity. The derivatives had significant but slower uptake rates in Staphylococcus aureus than [67Ga]-DFO (6% to 60% of the control rate), with no uptake for the most lipophilic derivatives. Biodistribution studies in mice with a S. aureus infection in one thigh revealed that the ethyl carbamate derivative had an excellent infected-to-non-infected ratio (11:1), but high non-specific localization in the gall bladder, liver and small intestine. CONCLUSIONS: The work reported shows that it is possible to functionalize DFO-type siderophores and retain active uptake of the 67Ga-labeled complexes by bacteria. Novel 67Ga-labeled DFO derivatives were specifically taken up by S. aureus and selected derivatives demonstrated in vivo localization at sites of infection. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: 67Ga-labeled DFO derivatives were actively transported by bacteria using the iron-siderophore pathway, suggesting that it is possible to develop siderophore-based radiopharmaceuticals for imaging bacterial infection.


Asunto(s)
Deferoxamina/química , Diagnóstico por Imagen/métodos , Radioisótopos de Galio/química , Sideróforos/química , Infecciones Estafilocócicas/diagnóstico por imagen , Animales , Transporte Biológico , Deferoxamina/metabolismo , Deferoxamina/farmacocinética , Femenino , Marcaje Isotópico , Ratones , Radioquímica , Sideróforos/metabolismo , Sideróforos/farmacocinética , Staphylococcus aureus/fisiología , Distribución Tisular
16.
Biomaterials ; 102: 58-71, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27322959

RESUMEN

Desferrioxamine (DFO) is currently in clinical use to remove iron from transfusion-dependent patients with ß-thalassemia major, sickle-cell anemia and the myelodysplastic syndromes. However, its short half-life, burdensome, subcutaneous mode of administration and propensity to cause neurotoxicity at high doses greatly hinder its use. Thus, developing an optimized version of DFO with extended half-life, and reduced toxicity is a major goal. Using high molecular weight (MW), non-toxic, hyperbranched polyglycerol with high functionality, we demonstrate that the efficacy of DFO can be tuned with considerable reduction in toxicity. Using zebrafish embryos and mice, we tested toxicity, iron removal efficacy with low dosing and the biodistribution of ultra-long circulating DFO (ULC-DFO) conjugates. There was no significant difference in the mortality and development of zebrafish embryos upon exposure to ULC-DFO. Similarly, body weights and serum lactate dehydrogenase levels in mice treated with ULC-DFO remained within the normal range throughout the tolerance study. Moreover, ULC-DFO is significantly more effective than low MW DFO in promoting iron removal both from organs and via urine in iron overloaded mice despite using a moderate, once-weekly dosing schedule. This is probably due to the extended circulation half-life of ULC-DFO. The MW of ULC-DFO influences the accumulation and biodistribution, with highest MW (637 KDa) associated with up to 12% accumulation in the liver. In contrast, ULC-DFO with MWs of 75 KDa and lower were associated with relatively low organ accumulation, indicating that biodistribution of ULC-DFO can be tuned. Since ULC-DFO has improved iron removal properties, longer plasma retention time and possesses excellent biocompatibility, it represents a polymer conjugate with high clinical utility in comparison to DFO for the treatment of transfusion dependent iron overload. More importantly, ULC-DFO is anticipated to reduce the requirement for prolonged subcutaneous infusion of DFO.


Asunto(s)
Deferoxamina/farmacocinética , Glicerol/farmacocinética , Quelantes del Hierro/farmacocinética , Polímeros/farmacocinética , Animales , Deferoxamina/química , Deferoxamina/uso terapéutico , Deferoxamina/toxicidad , Femenino , Glicerol/química , Glicerol/uso terapéutico , Glicerol/toxicidad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Quelantes del Hierro/química , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/toxicidad , Sobrecarga de Hierro/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Polímeros/química , Polímeros/uso terapéutico , Polímeros/toxicidad , Distribución Tisular , Pez Cebra
17.
J Control Release ; 235: 63-71, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27235979

RESUMEN

Poly(2-oxazoline)s are a promising class of polymers for biomedical applications and a versatile alternative to poly(ethylene glycol)s (PEG). In this work, the pharmacokinetic behavior of well defined (89)Zr-labeled poly(2-ethyl-2-oxazoline)s (PEtOx) was evaluated and compared to that of (89)Zr-labeled PEG, both with varying molar mass. Amine-terminated PEtOx of low dispersity in a molar mass range of 20 to 110kDa and PEG of 20 and 40kDa were functionalized with a desferrioxamine chelator and radiolabeled with (89)Zr. The tissue distribution of both radiolabeled PEtOx and PEG polymers was studied by means of micro Positron Emission Tomography (µPET) molecular imaging in mice longitudinally up to 1week post injection, followed by ex vivo biodistribution. As previously described for other classes of non-ionic polymers, the blood clearance of PEtOx decreased with molar mass. The cut off for glomerular filtration of PEtOx is likely to be around 40kDa. The head to head comparison of PEG and PEtOx revealed that the biodistribution is mostly dominated by polymer chain length and not polymer molar mass. This study constitutes an important addition to further establishing PEtOx as a promising polymer in biomedical applications.


Asunto(s)
Poliaminas/química , Poliaminas/farmacocinética , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Animales , Deferoxamina/administración & dosificación , Deferoxamina/química , Deferoxamina/farmacocinética , Isotiocianatos/química , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Peso Molecular , Miocardio/metabolismo , Tomografía de Emisión de Positrones/métodos , Radioisótopos , Sideróforos/administración & dosificación , Sideróforos/química , Sideróforos/farmacocinética , Distribución Tisular , Circonio
18.
J Nucl Med ; 57(3): 453-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26471693

RESUMEN

UNLABELLED: 5B1 is a fully human, monoclonal antibody that has shown promise for the PET imaging of cancers expressing carbohydrate antigen 19.9 (CA19.9)--a carbohydrate prevalent in cells with aberrant glycosylation and an established effector of metastasis. The long physiologic half-life of the antibody and interference from circulating CA19.9 may increase the time required to generate quality images as well as the risk of radiation exposure to healthy tissues during repeated PET imaging. Pretargeting methodologies are an effective approach to expeditiously acquire PET images, but in this case, the pretargeting approach is complicated by the internalization of 5B1 by CA19.9-expressing cells. We sought to adapt and optimize a pretargeting strategy that exploits the bioorthogonal reaction between transcyclooctene (TCO) and tetrazine (Tz) to overcome these complications. METHODS: 5B1 was modified with TCO, and a novel NOTA-PEG7-Tz radioligand was synthesized with the goal of improving on a previously reported analog. BxPC3 and Capan-2 cells were evaluated for their ability to internalize anti-CA19.9 antibodies using a fluorometric assay, and xenografts of the same lines were used for in vivo studies. The pretargeting approach was optimized, and the 2 radioligands were compared using biodistribution and PET imaging in murine models of pancreatic cancer. RESULTS: BxPC3 and Capan-2 cells were shown to rapidly internalize anti-CA19.9 monoclonal antibodies, including 5B1. (64)Cu-NOTA-PEG7-Tz showed improved in vivo pharmacokinetics relative to (64)Cu-NOTA-Tz using 5B1-TCO as the targeting vector. PET imaging and biodistribution studies showed that injecting the radioligand 72 h after the administration of 5B1-TCO resulted in the best uptake (8.2 ± 1.7 percentage injected dose per gram at 20 h after injection) and tumor-to-background activity concentration ratios. Dosimetry calculations revealed that the pretargeting system produced a greater than 25-fold reduction in total body radiation exposure relative to (89)Zr-desferrioxamine-5B1. PET/CT imaging in an orthotopic Capan-2 xenograft model--which secretes large amounts of CA19.9 and more rapidly internalizes anti-CA19.9 antibodies--showed that this approach is viable even in the difficult circumstances presented by a circulating antigen and internalized targeting vector. CONCLUSION: The 5B1-TCO and (64)Cu-NOTA-PEG7-Tz system evaluated in these studies can delineate CA19.9-positive xenografts in murine models of pancreatic cancer despite the challenges posed by the combination of circulating antigen and internalization of the 5B1-TCO.


Asunto(s)
Anticuerpos Antineoplásicos , Antígenos de Neoplasias , Neoplasias Pancreáticas/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Radioisótopos de Cobre , Deferoxamina/síntesis química , Deferoxamina/farmacocinética , Compuestos Heterocíclicos con 1 Anillo/síntesis química , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Humanos , Ratones , Trasplante de Neoplasias , Dosis de Radiación , Radioisótopos , Radiofármacos/síntesis química , Distribución Tisular , Circonio
19.
Bioconjug Chem ; 26(12): 2579-91, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26550847

RESUMEN

Zirconium-89 has an ideal half-life for use in antibody-based PET imaging; however, when used with the chelator DFO, there is an accumulation of radioactivity in the bone, suggesting that the (89)Zr(4+) cation is being released in vivo. Therefore, a more robust chelator for (89)Zr could reduce the in vivo release and the dose to nontarget tissues. Evaluation of the ligand 3,4,3-(LI-1,2-HOPO) demonstrated efficient binding of (89)Zr(4+) and high stability; therefore, we developed a bifunctional derivative, p-SCN-Bn-HOPO, for conjugation to an antibody. A Zr-HOPO crystal structure was obtained showing that the Zr is fully coordinated by the octadentate HOPO ligand, as expected, forming a stable complex. p-SCN-Bn-HOPO was synthesized through a novel pathway. Both p-SCN-Bn-HOPO and p-SCN-Bn-DFO were conjugated to trastuzumab and radiolabeled with (89)Zr. Both complexes labeled efficiently and achieved specific activities of approximately 2 mCi/mg. PET imaging studies in nude mice with BT474 tumors (n = 4) showed good tumor uptake for both compounds, but with a marked decrease in bone uptake for the (89)Zr-HOPO-trastuzumab images. Biodistribution data confirmed the lower bone activity, measuring 17.0%ID/g in the bone at 336 h for (89)Zr-DFO-trastuzumab while (89)Zr-HOPO-trastuzumab only had 2.4%ID/g. We successfully synthesized p-SCN-Bn-HOPO, a bifunctional derivative of 3,4,3-(LI-1,2-HOPO) as a potential chelator for (89)Zr. In vivo studies demonstrate the successful use of (89)Zr-HOPO-trastuzumab to image BT474 breast cancer with low background, good tumor to organ contrast, and, importantly, very low bone uptake. The reduced bone uptake seen with (89)Zr-HOPO-trastuzumab suggests superior stability of the (89)Zr-HOPO complex.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Mama/diagnóstico por imagen , Quelantes/química , Deferoxamina/química , Inmunoconjugados/química , Tomografía de Emisión de Positrones/métodos , Piridonas/química , Circonio/química , Animales , Línea Celular Tumoral , Quelantes/farmacocinética , Deferoxamina/farmacocinética , Femenino , Humanos , Inmunoconjugados/farmacocinética , Ratones Desnudos , Modelos Moleculares , Piridonas/farmacocinética , Distribución Tisular , Trastuzumab/química , Circonio/farmacocinética
20.
Bioconjug Chem ; 26(6): 1061-9, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-25971846

RESUMEN

The iron chelator deferoxamine (DFO), approved for the treatment of iron overload, has been examined as a therapeutic in a variety of conditions which iron may exacerbate. To evaluate the potential of DFO-bearing PEG-like nanoprobes (DFO-PNs) as therapeutics, we determined their pharmacokinetics (PK) in normal mice, and imaged their accumulation in a tumor model and in models of transient brain ischemia and inflammation. DFO-PNs consist of a DFO, a Cy5.5, and PEG (5 kDa or 30 kDa) attached to Lys-Cys scaffold. Tumor uptake of a [(89)Zr]:DFO-PN(10) (30 kDa PEG, diameter 10 nm) was imaged by PET, surface fluorescence, and fluorescence microscopy. DFO-PN(10) was internalized by tumor cells (fluorescence microscopy) and by cultured cells (by FACS). [(89)Zr]:DFO-PN(4.3) (5 kDa PEG, diameter 4.3 nm) concentrated at incision generated inflammations but not at sites of transient brain ischemia. DFO-PNs are fluorescent, PK tunable forms of DFO that might be investigated as antitumor or anti-inflammatory agents.


Asunto(s)
Isquemia Encefálica/diagnóstico , Deferoxamina/farmacocinética , Inflamación/diagnóstico , Quelantes del Hierro/farmacocinética , Neoplasias/diagnóstico , Animales , Encéfalo/patología , Carbocianinas/química , Carbocianinas/farmacocinética , Línea Celular Tumoral , Deferoxamina/química , Femenino , Quelantes del Hierro/química , Masculino , Ratones , Ratones Desnudos , Nanoestructuras/análisis , Nanoestructuras/química , Imagen Óptica , Polietilenglicoles/farmacocinética , Tomografía de Emisión de Positrones , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...