Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Photochem Photobiol B ; 199: 111630, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31610429

RESUMEN

Cancer disease is a foremost health concern and top basis of death in comparison with many diseases including cardiovascular disorders. During initial diagnosis (usually late diagnosis), a majority of cancer patients suffer from metastatic and advanced cancer stages which resulted in limited therapeutic modalities based interventions and effectiveness. Though considerable advancement has been made in combating the disease, continuous and intense efforts are ongoing for early diagnosis and development of therapies. Generally applied treatment options for cancer are surgery, chemotherapy and radiotherapy, which are restricted by failure to early diagnose, insufficient on-targeted drug delivery, systemic toxicity, and lack of real-time monitoring of therapeutic responses in cancer. Noninvasive imaging or minimally invasive imaging methodology is valuable in clinical diagnostic settings. Specifically, noninvasive optical imaging integrated with polymeric nanomaterial have been extensively investigated in the field of cancer diagnostics and therapy. Currently, optical imaging methods go together with polymer-based fluorescent nanoparticles in accomplishing the molecular level detection of tumor boundaries. NIR probe tagged polymeric nanoparticles have potential to provide an advantage in the early cancer detection, therapeutic monitoring and image guided surgery procedures. This article review the recent progress in state-of-the-art NIRF polymeric nanoparticles used for optical imaging particularly on cancer diagnosis.


Asunto(s)
Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Imagen Óptica/métodos , Polímeros/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Materiales Biocompatibles/química , Sistemas de Liberación de Medicamentos/métodos , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Humanos , Rayos Infrarrojos , Terapia Molecular Dirigida/métodos , Fosfolípidos/química , Propiedades de Superficie
2.
Ultrasound Med Biol ; 44(4): 909-914, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29395679

RESUMEN

Chronic wounds typically require long-duration treatment with a combination of antibiotics administered systemically. This incurs adverse side effects and can require aversive surgical treatments and limb amputations. To improve non-invasive antimicrobial therapy, the objective of this study was to investigate antimicrobial chemotherapy combined with high-intensity focused ultrasound (HIFU) heating (HT). A Staphylococcus aureus abscess (80 ± 30 mm3) was generated in the mouse flank region. Once the average temperature (~42 °C-46 °C) in the abscess was reached with HIFU-HT, a broad-spectrum antimicrobial (ciprofloxacin, 10 mg/kg) and perfusion marker (Evans blue dye, 40 mg/kg wt) were administered intravenously via the tail vein. Four hours later, mean abscess perfusion and colony-forming units (CFUs) per gram of abscess were determined. HIFU-HT increased abscess perfusion by ~2.5-fold (4 ± 0.6 µg/mL Evans blue) compared with control (1.5 ± 0.7 µg/mL), and improved antimicrobial efficacy to decrease percentage average survival of S. aureus by ~20% (46 ± 7 CFUs/g of abscess) versus that seen with ciprofloxacin alone (61 ± 4 CFU/g). Our in vivo data suggest that HIFU-HT can improve antimicrobial treatment responses against deep-seated bacteria in abscess wounds via enhanced perfusion.


Asunto(s)
Absceso/tratamiento farmacológico , Antibacterianos/uso terapéutico , Ciprofloxacina/uso terapéutico , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus , Animales , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Calor , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
3.
Mol Pharm ; 14(7): 2224-2235, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28505457

RESUMEN

Red blood cells (RBCs) express a variety of immunomodulatory markers that enable the body to recognize them as self. We have shown that RBC membrane glycophorin A (GPA) receptor can mediate membrane attachment of protein therapeutics. A critical knowledge gap is whether attaching drug-encapsulated nanoparticles (NPs) to GPA and modification with cell-penetrating peptide (CPP) will impact binding, oxygenation, and the induction of cellular stress. The objective of this study was to formulate copolymer-based NPs containing model fluorescent-tagged bovine serum albumin (BSA) with GPA-specific targeting ligands such as ERY1 (ENPs), single-chain variable antibody (scFv TER-119, SNPs), and low-molecular-weight protamine-based CPP (LNPs) and to determine their biocompatibility using a variety of complementary high-throughput in vitro assays. Experiments were conducted by coincubating NPs with RBCs at body temperature, and biocompatibility was evaluated by Raman spectroscopy, hemolysis, complement lysis, and oxidative stress assays. Data suggested that LNPs effectively targeted RBCs, conferring 2-fold greater uptake in RBCs compared to ENPs and SNPs. Raman spectroscopy results indicated no adverse effect of NP attachment or internalization on the oxygenation status of RBCs. Cellular stress markers such as glutathione, malondialdehyde, and catalase were within normal limits, and complement-mediated lysis due to NPs was negligible in RBCs. Under the conditions tested, our data demonstrates that molecular targeting of the RBC membrane is a feasible translational strategy for improving drug pharmacokinetics and that the proposed high-throughput assays can prescreen diverse NPs for preclinical and clinical biocompatibility.


Asunto(s)
Péptidos de Penetración Celular/química , Nanopartículas/química , Polímeros/química , Animales , Bovinos , Péptidos de Penetración Celular/farmacología , Sistemas de Liberación de Medicamentos/métodos , Eritrocitos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Albúmina Sérica Bovina/química , Espectrometría Raman
4.
Pharm Res ; 33(5): 1191-203, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26812966

RESUMEN

PURPOSE: Nanoparticle (NP) attachment to biocompatible secondary carriers such as red blood cell (RBC) can prolong blood residence time of drug molecules and help create next-generation nanotherapeutics. However, little is known about the impact of RBC-targeted NPs on erythrocyte function. METHODS: The objectives of this study were to develop and characterize in vitro a novel poly-L-lysine (PLL) and polyethylene glycol (PEG) copolymer-based NP containing fluorescent-tagged bovine serum albumin (BSA), and conjugated with ERY1, a 12 amino acid peptide with high affinity for the RBC membrane protein glycophorin A (ENP). RESULTS: Confocal and flow cytometry data suggest that ENPs efficiently and irreversibly bind to RBC, with approximately 70% of erythrocytes bound after 24 h in a physiologic flow loop model compared to 10% binding of NPs without ERY1. Under these conditions, synthesized ENPs were not toxic to the RBCs. The rheological parameters at the applied shear. (0-15 Pa) were not influenced by ENP attachment to the RBCs. However, at high concentration, the strong affinity of ENPs to the glycophorin-A reduced the deformability of the RBC. CONCLUSIONS: ENPs can be efficiently attached to the RBCs without adversely affecting cellular function, and this may potentially enhance circulatory half-life of drug molecules.


Asunto(s)
Sistemas de Liberación de Medicamentos , Membrana Eritrocítica/metabolismo , Glicoforinas/metabolismo , Nanopartículas/metabolismo , Péptidos/metabolismo , Animales , Bovinos , Deformación Eritrocítica , Eritrocitos/citología , Eritrocitos/metabolismo , Ratones , Nanopartículas/química , Péptidos/química , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polilisina/química , Polilisina/metabolismo , Albúmina Sérica Bovina/administración & dosificación
5.
Int J Hyperthermia ; 31(6): 674-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26185910

RESUMEN

BACKGROUND/OBJECTIVE: Novel approaches allowing efficient, readily translatable image-guided drug delivery (IGDD) against solid tumours is needed. The objectives of this study were to: 1) develop echogenic low temperature sensitive liposomes (E-LTSLs) loaded with an ultrasound (US) contrast agent (perfluoropentane, PFP), 2) determine the in vitro and in vivo stability of contrast agent encapsulation, 3) co-encapsulate and characterise doxorubicin (Dox) E-LTSL, and cellular uptake and cytotoxicity in combination with high intensity focused ultrasound (HIFU). METHOD: E-LTSLs were loaded passively with PFP and actively with Dox. PFP encapsulation in E-LTSL was determined by transmission electron microscopy (TEM), and US imageability was determined in tissue-mimicking phantoms and mouse tumour model. Dox release from E-LTSL in physiological buffer was quantified by fluorescence spectroscopy. Cellular uptake and cytotoxicity of E-LTSL in the presence of HIFU-induced mild hyperthermia (∼40-42 °C) was determined in a 3D tumour spheroid model. RESULTS: TEM and US confirmed that the PFP emulsion was contained within LTSLs. Phantom and animal studies showed that the E-LTSLs were echogenic. Temperature versus size increase and Dox release kinetics of E-LTSLs demonstrated no difference compared to LTSL alone. Dox release was <5% within 1 h at baseline (25 °C) and body (37 °C) temperatures, and was >99% under hyperthermia. E-LTSL plus HIFU achieved significantly greater Dox uptake in spheroids and cytotoxicity compared to body temperature. CONCLUSION: A stable US-imageable liposome co-loaded with Dox and PFP for in vivo IGDD was developed. Data suggest that HIFU can induce cellular uptake and toxicity with E-LTSLs.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Medios de Contraste/administración & dosificación , Doxorrubicina/administración & dosificación , Fluorocarburos/administración & dosificación , Ultrasonido Enfocado de Alta Intensidad de Ablación , Neoplasias/terapia , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Medios de Contraste/farmacocinética , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Fluorocarburos/farmacocinética , Calor , Humanos , Liposomas , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
6.
J Pharmacol Exp Ther ; 351(2): 413-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25204337

RESUMEN

Gut is very sensitive to hypoperfusion and hypoxia, and deranged gastrointestinal barrier is implicated in systemic failure of various organs. We recently demonstrated that diphenyldihaloketone EF24 [3,5-bis(2-fluorobenzylidene)piperidin-4-one] improves survival in a rat model of hemorrhagic shock. In this study, we tested EF24 and its other analog CLEFMA (4-[3,5-bis(2-chlorobenzylidene)-4-oxo-piperidine-1-yl]-4-oxo-2-butenoic acid) for their effect on intestinal barrier dysfunction in hypovolemic shock. Hypovolemia was induced in rats by withdrawing 50% of blood. EF24 or CLEFMA (0.4 mg/kg i.p.) treatment was provided, without volume resuscitation, after 1 hour of hemorrhage. Ileum was collected 5 hours after the treatment to investigate the expression of tight junction proteins (zonula occludens, claudin, and occludin) and epithelial injury markers [myeloperoxidase, ileal lipid-binding protein (ILBP), CD163, and plasma citrulline]. The ileal permeability for dextran-fluoroisothiocyanate and Evan's blue dye was determined. EF24 and CLEFMA reduced the hypovolemia-induced plasma citrulline levels and the ileal expression of myeloperoxidase, ILBP, and CD163. The drugs also restored the basal expression levels of zonula occludens, claudin, and occludin, which were substantially deranged by hypovolemia. In ischemic ileum, the expression of phospho(tyrosine)-zonula occludens-1 was reduced, which was reinstated by EF24 and CLEFMA. In contrast, the drug treatments maintained the hypovolemia-induced expression of phospho(threonine)-occludin, but reduced that of phospho(tyrosine)-occludin. Both EF24 and CLEFMA treatments reduced the intestinal permeability enhanced by hypovolemia. EF24 and CLEFMA attenuate hypovolemic gut pathology and protect barrier function by restoring the status of tight junction proteins. These effects were observed in unresuscitated shock, implying the benefit of EF24 and CLEFMA in prehospital care of shock.


Asunto(s)
Compuestos de Bencilideno/farmacología , Íleon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Piperidonas/farmacología , Choque Hemorrágico/tratamiento farmacológico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Íleon/metabolismo , Íleon/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Permeabilidad/efectos de los fármacos , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patología , Proteínas de Uniones Estrechas/metabolismo
7.
Br J Pharmacol ; 170(7): 1436-48, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24102070

RESUMEN

BACKGROUND AND PURPOSE: 4-[3,5-Bis(2-chlorobenzylidene)-4-oxo-piperidine-1-yl]-4-oxo-2-butenoic acid CLEFMA is a new anti-cancer molecule. Here, we investigated changes in apoptosis and inflammatory markers during CLEFMA-induced tumour suppression. EXPERIMENTAL APPROACH: Lung adenocarcinoma H441 and A549, and normal lung fibroblast CCL151 cell lines were used, along with a xenograft model of H441 cells implanted in mice. Tumour tissues were analysed by immunoblotting, immunohistochemistry and/or biochemical assays. The ex vivo results were confirmed by performing selected assays in cultured cells. KEY RESULTS: CLEFMA-induced cell death was associated with cleavage of caspases 3/9 and PARP. In vivo, CLEFMA treatment resulted in a dose-dependent suppression of tumour growth and (18) F-fluorodeoxyglucose uptake in tumours, along with a reduction in the expression of the proliferation marker Ki-67. In tumour tissue homogenates, the anti-apoptotic markers (cellular inhibitor of apoptosis protein-1(cIAP1), Bcl-xL, Bcl-2, and survivin) were inhibited and the pro-apoptotic Bax and BID were up-regulated. Further, CLEFMA decreased translocation of phospho-p65-NF-κB into the nucleus. In vitro, it inhibited the DNA-binding and transcriptional activity of NF-κB. It also reduced the expression of COX-2 in tumours and significantly depressed serum TNF-α and IL-6 levels. These effects of CLEFMA were accompanied by a reduced transcription and/or translation of the invasion markers VEGF, MMP9, MMP10, Cyclin D1 and ICAM-1. CONCLUSIONS AND IMPLICATIONS: Overall, CLEFMA inhibited growth of lung cancer xenografts and this tumour suppression was associated with NF-κB-regulated anti-inflammatory and anti-metastatic effects.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Compuestos de Bencilideno/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Piperidonas/farmacología , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Fluorodesoxiglucosa F18 , Regulación Neoplásica de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Imagen Multimodal , Fosforilación , Tomografía de Emisión de Positrones , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Tomografía Computarizada por Rayos X , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pharmacol Exp Ther ; 347(2): 346-56, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23995597

RESUMEN

An exaggerated release of proinflammatory cytokines and accompanying inflammation contributes to the development of multiple organ failure after hemorrhagic shock. Here, we tested the nuclear factor (NF) κ-light-chain-enhancer of activated B cell (NF-κB)-mediated transcriptional control of inflammatory pathways as a target in the management of hemorrhage-induced inflammation. We performed a study in a rat model of fixed-volume hemorrhage to investigate the anti-inflammatory effects of the diphenyldifluoroketone EF24 [3,5-bis(2-fluorobenzylidene)piperidin-4-one], an NF-κB inhibitor, in lung tissue. EF24 treatment (0.4 mg/kg) significantly prevented the upregulation of inflammatory biomarkers in rats subjected to 50% hemorrhage and preserved the pulmonary histology in hemorrhaged rats. The lung tissue from treated rats showed marked suppression of the hemorrhage-mediated induction of Toll-like receptor 4, phospho-p65 NF-κB, inducible nitric-oxide synthase, heme oxygenase-1, and cyclooxygenase-2 (COX-2). The hemorrhage-induced COX-2 activity was also significantly inhibited by the EF24 treatment. At the same time, EF24 induced nuclear factor (erythroid-derived 2)-like 2-mediated protective mechanisms against oxidative stress. EF24 also reduced hemorrhage-induced lung myeloperoxidase activity. The plasma levels of proinflammatory tumor necrosis factor-α, interleukin (IL)-6, IL-1α, and IL-1ß were lower in EF24-treated rats than in untreated rats. Moreover, there was a significant reduction in the pulmonary expression of high-mobility group B1 protein. These biochemical effects were accompanied by a significant improvement in the survival of rats administered with EF24 as compared with the rats receiving vehicle control (P < 0.05). Overall, the results suggest that EF24 attenuates hemorrhage-induced inflammation and could serve as a salutary anti-inflammatory agent in resuscitation strategies.


Asunto(s)
Antiinflamatorios/uso terapéutico , Compuestos de Bencilideno/uso terapéutico , Hipovolemia/prevención & control , Piperidonas/uso terapéutico , Choque Hemorrágico/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Compuestos de Bencilideno/administración & dosificación , Compuestos de Bencilideno/farmacología , Ciclooxigenasa 2/biosíntesis , Citocinas/sangre , Citocinas/inmunología , Modelos Animales de Enfermedad , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hipovolemia/etiología , Hipovolemia/inmunología , Hipovolemia/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Piperidonas/administración & dosificación , Piperidonas/farmacología , Ratas , Ratas Sprague-Dawley , Choque Hemorrágico/complicaciones , Choque Hemorrágico/inmunología , Choque Hemorrágico/patología , Análisis de Supervivencia , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/biosíntesis
9.
Invest New Drugs ; 30(2): 558-67, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21181232

RESUMEN

CLEFMA or 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] is a curcuminoid being developed as an anticancer drug. We recently reported that it potently inhibits proliferation of various cancer cells. In this project, we investigated the effect of CLEFMA on gene expression profile in H441 lung adenocarcinoma cells, and studied its mechanism of action. In microarray data, we observed a deregulation of genes involved in redox and glutamate metabolism. Based on the affected ontologies, we hypothesized that antiproliferative activity of CLEFMA could be a result of the induction of reactive oxygen species (ROS). We tested this hypothesis by determining the levels of glutathione (GSH) and ROS in H441 cells treated with CLEFMA. We observed a rapid depletion of intracellular GSH/GSSG ratio. Using a cell-permeable fluorogenic substrate, we found that CLEFMA significantly induced ROS in a time- and dose-dependent manner (p < 0.05). Flow-cytometry with a mitochondria-selective fluorescent reporter of ROS indicated that the CLEFMA-induced ROS was of mitochondrial origin. In contrast to the cancer cells, the normal lung fibroblasts (CCL-151) did not show any increase in ROS and were resistant to CLEFMA-induced cell death. Furthermore, the addition of antioxidants, such as catalase, superoxide dismutase and N-acetylcysteine, rescued cancer cells from CLEFMA-induced cell death. Gene expression pathway analysis suggested that a transcription factor regulator Nrf2 is a pivotal molecule in the CLEFMA-induced deregulation of redox pathways. The immunoblotting of Nrf2 showed that CLEFMA treatment resulted in phosphorylation and nuclear translocation of Nrf2 in a time-dependent fashion. Based on these results, we conclude that induction of ROS is critical for the antiproliferative activity of CLEFMA and the Nrf2-mediated oxidative stress response fails to salvage H441 cells.


Asunto(s)
Adenocarcinoma/metabolismo , Antineoplásicos Fitogénicos/farmacología , Compuestos de Bencilideno/farmacología , Curcumina , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Piperidonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transporte Activo de Núcleo Celular , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antioxidantes/farmacología , Western Blotting , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción , Estrés Oxidativo/genética , Fosforilación , Factores de Tiempo
10.
Chem Biol Drug Des ; 79(2): 194-201, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22107757

RESUMEN

3,5-Bis(2-fluorobenzylidine)-4-piperidone or EF24 is a potent anticancer derivative of curcumin. Using an amine derivative of EF24, we synthesized a hydrazinonicotinic acid conjugate, EFAH, for Tc-99m radiolabelling and single photon emission tomography imaging. The aqueous solubility of EFAH (3.5 mg/mL) was significantly more than that of EF24 (1.2 mg/mL); the octanol/water partition coefficient of EFAH was estimated at log P = 0.33. As an antiproliferative agent, EFAH was as effective as EF24 in suppressing the proliferation of H441, MiaPaCa-2 and Panc-1 cells. Daily intraperitoneal injection of EFAH (5 µg) for 3 weeks in mice carrying xenografts of Panc-1 pancreatic cancer showed a mean tumour volume reduction of 79%; the tumour weight decreased by 82% in the treated group. For imaging and biodistribution, EFAH was labelled with Tc-99m (98% RCY) and intravenously administered in rats. Approximately 23.7% and 14.3% of injected dose accumulated in liver and intestine, respectively, suggesting that EFAH is mostly eliminated by hepatobiliary route. The results indicate that HYNIC modification of EF24 for Tc-99m radiolabelling does not affect its antiproliferative efficacy. For the first time, a visual biodisposition of EF24 in a live animal model has been demonstrated. Such knowledge could be of benefit in developing therapeutic curcuminoids, such as EF24.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bencilideno/química , Hidrazinas/farmacología , Piperidonas/química , Piperidonas/farmacología , Radiofármacos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Hidrazinas/síntesis química , Hidrazinas/química , Hidrazinas/farmacocinética , Marcaje Isotópico , Ratones , Ratones Desnudos , Piperidonas/síntesis química , Piperidonas/farmacocinética , Radiofármacos/síntesis química , Radiofármacos/química , Radiofármacos/farmacocinética , Ratas , Tecnecio/química , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Trasplante Heterólogo
11.
Nucl Med Commun ; 32(10): 908-12, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21876402

RESUMEN

OBJECTIVE: We recently identified p-[(18)F]fluorohippurate ([(18)F]PFH) as a potential positron emission tomography (PET) renal agent. The objective of this study was to compare renogram parameters of [(18)F]PFH with o-[(125)I]iodohippurate ([(125)I]OIH) as a surrogate for the renal imaging gold standard (131)I-OIH and with a clinically used agent [(99m)Tc]MAG3. METHODS: Normal Sprague-Dawley rats (n=4) were sequentially imaged on days 1, 2, and 3 using [(99m)Tc]MAG3 (18.1-19.1 MBq, dynamic planar), [(18)F]PFH (2.6-4.1 MBq, dynamic PET), and [(125)I]OIH (7.7-13.5 MBq, dynamic planar), respectively. The PET data were binned into frames of 30 s each, whereas the planar images were acquired as 30 s per frame. Regions of interest were drawn on both kidneys, and decay-corrected renograms were generated for each imaging modality. RESULTS: The PET-derived [(18)F]PFH renograms revealed an average time-to-peak (T(max)) of 4.8±2.4 min, which was comparable to the T(max) of 3.6±1.7 min and 4.3±1.7 min for [(125)I]OIH and [(99m)Tc]MAG3 renograms, respectively. The average time-to-half-maximal activity was found to be 16.6±6.6 min, 8.3±2.4 min, and more than 20 min with [(18)F]PFH, [(125)I]OIH, and [(99m)Tc]MAG3, respectively. CONCLUSIONS: Compared with [(99m)Tc]MAG3, the renogram parameters of [(18)F]PFH seem to be closer to those obtained from [(125)I]OIH. The quality of the renogram and the images obtained with the dynamic [(18)F]PFH PET study were remarkably better than those obtained with the [(99m)Tc]MAG3 dynamic planar imaging study.


Asunto(s)
Radioisótopos de Flúor , Hipuratos , Ácido Yodohipúrico , Renografía por Radioisótopo/métodos , Tecnecio Tc 99m Mertiatida , Animales , Radioisótopos de Yodo , Túbulos Renales/diagnóstico por imagen , Túbulos Renales/fisiología , Renografía por Radioisótopo/normas , Ratas , Estándares de Referencia
12.
Colloids Surf B Biointerfaces ; 84(2): 329-37, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21315563

RESUMEN

We recently reported a novel curcuminoid 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] or CLEFMA as a potent anti-proliferative agent, and showed that it induces autophagic cell death in lung cancer cells. We are now reporting a drug-in-CD-in-liposome approach to formulate CLEFMA liposomes that could be labeled with Tc-99m radionuclide for non-invasive imaging of their biodistribution. CLEFMA encapsulation was enabled by hydroxypropyl-ß-cyclodextrin. In vitro studies showed that CLEFMA possessed more potent anti-proliferative activity in lung adenocarcinoma H441 cells than naturally occurring curcumin. At the same time, it had no effect on the proliferative capacity of normal lung fibroblasts. CLEFMA liposomes retained the antiproliferative potency of free CLEFMA, while maintaining its non-toxic nature in normal lung fibroblasts. In nude rats bearing xenograft H441 tumors, the tumor volume significantly reduced after i.v. treatment with CLEFMA liposomes (p<0.05); the tumor inhibition was determined to be 94%. The anti-tumor activity of CLEFMA liposomes was confirmed by the observation that F-18-fluorodeoxyglucose uptake in tumors of treated rats was reduced as compared to those of control rats. Tc-99m-labeled CLEFMA liposomes accumulated in liver (33.7%); spleen showed the largest accumulation on per gram tissue basis (6.2%/g). Upon histopathological examination of liver, lung and kidney, we found no apparent toxicity from multiple CLEFMA liposome administrations. The results demonstrate the utility of liposomes to serve as a carrier for CLEFMA. This study is the first to demonstrate the efficacy of novel curcuminoid CLEFMA in a preclinical model.


Asunto(s)
Ciclodextrinas/química , Sistemas de Liberación de Medicamentos , Liposomas , Neoplasias Pulmonares/tratamiento farmacológico , Piperidonas/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Modelos Animales de Enfermedad , Piperidonas/química , Ratas
13.
Bioorg Med Chem Lett ; 20(20): 5969-72, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20829039

RESUMEN

Two α(v)ß(3) integrin-binding peptide conjugates containing the cyclic CisoDGRC motif, a linker, and a chelator to enable Tc-99m labeling via the fac-[(99m)Tc(CO)(3)]+ core were synthesized. In vivo biodistribution studies in U87MG tumor-bear nude mice at 1h post-injection revealed a profound effect of the linker on the clearance of the radiotracer from the blood stream. In vivo blocking studies demonstrated the selective binding to the tumors expressing α(v)ß(3)-integrin and other tissues. The HPLC analysis of urine samples collected upon necropsy showed no degradation indicating their metabolic stability. These results suggest that cyclic CisoDGRC motif could be exploited as a new α(v)ß(3)-targeting vector by an appropriate selection of a linker between the peptide and the payload to obtain optimum pharmacokinetic properties.


Asunto(s)
Glioblastoma/diagnóstico por imagen , Integrina alfaVbeta3/análisis , Péptidos Cíclicos , Radiofármacos , Tecnecio , Tomografía Computarizada de Emisión de Fotón Único/métodos , Secuencias de Aminoácidos , Animales , Humanos , Integrina alfaVbeta3/metabolismo , Ratones , Ratones Desnudos , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Unión Proteica , Radiofármacos/síntesis química , Radiofármacos/química , Tecnecio/química , Distribución Tisular
14.
Bioorg Med Chem ; 18(16): 6109-20, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20638855

RESUMEN

3,5-Bis(benzylidene)-4-piperidones are being advanced as synthetic analogs of curcumin for anti-cancer and anti-inflammatory properties. We performed structure-activity relationship studies, by testing several synthesized 3,5-bis(benzylidene)-4-piperidones for anti-proliferative activity in lung adenocarcinoma H441 cells. Compared to the lead compound 1, or 3,5-bis(2-fluorobenzylidene)-4-piperidone, five compounds were found to be more potent (IC(50) < 30 microM), and 16 compounds possessed reduced cell-killing efficacy (IC(50) > 50 microM). Based on the observations, we synthesized 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] (29 or CLEFMA) as a novel analog of 1. CLEFMA was evaluated for anti-proliferative activity in H441 cells, and was found to be several folds more potent than compound 1. We did not find apoptotic cell population in flow cytometry, and the absence of apoptosis was confirmed by the lack of caspase cleavage. The electron microscopy of H441cells indicated that CLEFMA and compound 1 induce autophagic cell death that was inhibited by specific autophagy inhibitor 3-methyladenine. The results suggest that the potent and novel curcuminoid, CLEFMA, offers an alternative mode of cell death in apoptosis-resistant cancers.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos de Bencilideno/química , Compuestos de Bencilideno/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Piperidonas/química , Piperidonas/farmacología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Humanos , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA