Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830136

RESUMEN

The selection of technological parameters for nanoparticle formulation represents a complicated development phase. Therefore, the statistical analysis based on Box-Behnken methodology is widely used to optimize technological processes, including poly(lactic-co-glycolic acid) nanoparticle formulation. In this study, we applied a two-level three-factor design to optimize the preparation of nanoparticles loaded with cobalt (CoTPP), manganese (MnClTPP), and nickel (NiTPP) metalloporphyrins (MeP). The resulting nanoparticles were examined by dynamic light scattering, X-ray diffraction, Fourier transform infrared spectroscopy, MTT test, and hemolytic activity assay. The optimized model of nanoparticle formulation was validated, and the obtained nanoparticles possessed a spherical shape and physicochemical characteristics enabling them to deliver MeP in cancer cells. In vitro hemolysis assay revealed high safety of the formulated MeP-loaded nanoparticles. The MeP release demonstrated a biphasic profile and release mechanism via Fick diffusion, according to release exponent values. Formulated MeP-loaded nanoparticles revealed significant antitumor activity and ability to generate reactive oxygen species. MnClTPP- and CoTPP-nanoparticles specifically accumulated in tissues, preventing wide tissue distribution caused by long-term circulation of the hydrophobic drug. Our results suggest that MnClTPP- and CoTPP-nanoparticles represent the greatest potential for utilization in in anticancer therapy due to their effectiveness and safety.


Asunto(s)
Complejos de Coordinación/farmacocinética , Metaloporfirinas/farmacocinética , Metales/química , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Porfirinas/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Liberación de Fármacos , Femenino , Células HeLa , Hemólisis/efectos de los fármacos , Humanos , Células MCF-7 , Metaloporfirinas/química , Metaloporfirinas/farmacología , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Nanopartículas/ultraestructura , Ratas Wistar , Espectroscopía Infrarroja por Transformada de Fourier , Distribución Tisular , Difracción de Rayos X
2.
J Pharmacol Exp Ther ; 377(1): 1-10, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33500265

RESUMEN

Oxidative stress plays a crucial role in the pathogenesis of Parkinson disease (PD), and one strategy for neuroprotective therapy for PD is to scavenge reactive species using a catalytic antioxidant. Previous studies in our laboratory revealed that pretreatment of lipophilic metalloporphyrins showed protective effects in a mouse PD model. In this study, we optimized the formulations of these metalloporphyrins to deliver them orally and tested their efficacy on disease outcomes in a second species after initiation of an insult (i.e., disease modification). In this study, a pharmaceutical formulation of two metalloporphyrin catalytic antioxidants, AEOL11207 and AEOL11114, was tested for oral drug delivery. Both compounds showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier after intravenous or oral delivery. AEOL11207 and AEOL11114 bioavailabilities were calculated to be 24% and 25%, respectively, at a dose of 10 mg/kg via the oral route. In addition, both compounds significantly attenuated 6-hydroxydopamine (6-OHDA)-induced neurotoxic damage, including dopamine depletion, cytokine production, and microglial activation in the striata; dopaminergic neuronal loss in the substantia nigra; oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain; and rotation behavioral abnormality in rats. These results indicate that AEOL11207 and AEOL11114 are orally active metalloporphyrins and protect against 6-OHDA neurotoxicity 1-3 days postlesioning, suggesting disease-modifying properties and translational potential for PD. SIGNIFICANCE STATEMENT: Two catalytic antioxidants showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier. Both compounds significantly attenuated dopamine depletion, cytokine production, microglial activation, dopaminergic neuronal loss, oxidative/nitrative stress indices, and behavioral abnormality in a Parkinson disease rat model. The results suggest that both metalloporphyrins possess disease-modifying properties that may be useful in treating Parkinson disease.


Asunto(s)
Antioxidantes/farmacocinética , Metaloporfirinas/farmacocinética , Fármacos Neuroprotectores/farmacocinética , Trastornos Parkinsonianos/tratamiento farmacológico , Administración Oral , Animales , Antioxidantes/administración & dosificación , Antioxidantes/uso terapéutico , Barrera Hematoencefálica/metabolismo , Masculino , Metaloporfirinas/administración & dosificación , Metaloporfirinas/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Distribución Tisular
3.
Proc Natl Acad Sci U S A ; 117(13): 7021-7029, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32179677

RESUMEN

Described here is the development of gadolinium(III) texaphyrin-platinum(IV) conjugates capable of overcoming platinum resistance by 1) localizing to solid tumors, 2) promoting enhanced cancer cell uptake, and 3) reactivating p53 in platinum-resistant models. Side by side comparative studies of these Pt(IV) conjugates to clinically approved platinum(II) agents and previously reported platinum(II)-texaphyrin conjugates demonstrate that the present Pt(IV) conjugates are more stable against hydrolysis and nucleophilic attack. Moreover, they display high potent antiproliferative activity in vitro against human and mouse cell cancer lines. Relative to the current platinum clinical standard of care (SOC), a lead Gd(III) texaphyrin-Pt(IV) prodrug conjugate emerging from this development effort was found to be more efficacious in subcutaneous (s.c.) mouse models involving both cell-derived xenografts and platinum-resistant patient-derived xenografts. Comparative pathology studies in mice treated with equimolar doses of the lead Gd texaphyrin-Pt(IV) conjugate or the US Food and Drug Administration (FDA)-approved agent oxaliplatin revealed that the conjugate was better tolerated. Specifically, the lead could be dosed at more than three times (i.e., 70 mg/kg per dose) the tolerable dose of oxaliplatin (i.e., 4 to 6 mg/kg per dose depending on the animal model) with little to no observable adverse effects. A combination of tumor localization, redox cycling, and reversible protein binding is invoked to explain the relatively increased tolerability and enhanced anticancer activity seen in vivo. On the basis of the present studies, we conclude that metallotexaphyrin-Pt conjugates may have substantial clinical potential as antitumor agents.


Asunto(s)
Antineoplásicos/administración & dosificación , Metaloporfirinas/administración & dosificación , Oxaliplatino/administración & dosificación , Células A549 , Animales , Antineoplásicos/farmacocinética , Resistencia a Antineoplásicos , Femenino , Células HCT116 , Humanos , Metaloporfirinas/farmacocinética , Ratones Desnudos , Oxaliplatino/farmacocinética , Profármacos/administración & dosificación , Profármacos/farmacocinética , Distribución Tisular , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Int J Toxicol ; 38(4): 291-302, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333066

RESUMEN

Since our earlier publication (Gad et al, 2013), BioMimetix has advanced BMX-010 (Manganese (III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin or MnTE020PyP; CASRN 219818-60-7) into clinical development as a topical agent for the treatment of psoriasis, atopic dermatitis, and pruritus (idiopathic nonspecific itch). A multiple dose phase I study has been completed in 64 patients without any serious adverse effects. During the course of development, the formulation was initially a gel but has been modified to a cream formulation. The nonclinical safety program has been carried onward to assess preclinical risk to patients. Additional studies completed and reported here include dermal sensitization in a Guinea Pig maximization test study, 2 rabbit phototoxicity studies, a 28-day oral toxicity study in juvenile mice, a 28-day topical systemic toxicity study in Gottingen minipigs, range-finding studies, and complete embryo-fetal developmental toxicity (Segment II) studies in mice and rabbits, an ICH M7 compliant qualification of impurities using 2 (Q)SAR in silico methods, and a 14-day subcutaneous toxicity study of mice to qualify an impurity. All studies (except the (Q)SAR evaluations) were performed in accordance with Good Laboratory Practices (GLP) using Good Manufacturing Practices (GMP) drug substance. The systemic toxicity studies, with the exception of the juvenile toxicity study, included toxicokinetic evaluations, which are reported here. The phase I clinical study had 67 patient participants who received topically applied BMX-010, and there were no notable safety findings and included pharmacokinetic determinations on these patients which are also reported here. Chronic GLP toxicity studies have been initiated in the mouse (6-month oral) and minipig (9-month dermal). To date, the only observed nonclinical toxicity remains a reversible hypertension seen in mice in response to Cmax levels with a no effect threshold, and there have been no drug-related adverse effects.


Asunto(s)
Metaloporfirinas/farmacocinética , Metaloporfirinas/toxicidad , Administración Cutánea , Animales , Dermatitis Atópica/tratamiento farmacológico , Femenino , Cobayas , Humanos , Hipertensión/inducido químicamente , Masculino , Ratones , Psoriasis/tratamiento farmacológico , Conejos , Porcinos , Porcinos Enanos , Pruebas de Toxicidad
5.
J Biomed Mater Res A ; 106(11): 2955-2962, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30260555

RESUMEN

To enhance anticancer efficacy and decrease side effects, the synergistic multi-agent therapy has now increasingly gotten great attention. Herein, the flexible polycaprolactone had been modified by histidine and formed assembles with PEGylated metallo-porphyrin via metal-coordinated supramolecular interaction. This supramolecular assembles showed excellent acid sensitivity. At neutral environment, the hydrophobic anticancer drug could be effectively co-encapsulated with photosensitizer Fe-TPP to improve the water solubility. While at the intracellular microenvironment, the changed acid environment would trigger the drug and Fe-TPP release due to the reduction of metal-coordinated interaction between histidine and metallo-porphyrin leading to the disintegration of assembles. The in vivo anticancer experiments toward HeLa and MCF-7 cells disclosed that this co-delivery system of anticancer drug and photosensitizer showed enhanced anticancer efficacy, implying that the synergistic chemo-photodynamic therapy could improve cellular proliferation inhibition. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2955-2962, 2018.


Asunto(s)
Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/química , Doxorrubicina/administración & dosificación , Metaloporfirinas/administración & dosificación , Fármacos Fotosensibilizantes/administración & dosificación , Poliésteres/química , Ácidos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Complejos de Coordinación/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Células HeLa , Histidina/análogos & derivados , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Metaloporfirinas/farmacología , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/farmacología , Polietilenglicoles/química
6.
Cancer Chemother Pharmacol ; 80(2): 421-431, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28685347

RESUMEN

PURPOSE: Manganese porphyrins are redox-active drugs and superoxide dismutase mimics, which have been shown to chemosensitize lymphoma, a cancer which frequently occurs in dogs. This study aimed to identify critical information regarding the pharmacokinetics and toxicity of Mn(III) meso-tetrakis (N-n-butoxyetylpyridium-2-yl) porphyrin, (MnTnBuOE-2-PyP5+, MnBuOE) in dogs as a prelude to a clinical trial in canine lymphoma patients. METHODS: A single-dose pharmacokinetic (PK) study in normal dogs was performed to determine the plasma half-life (t 1/2) of MnBuOE. A dose reduction study was performed to establish the maximum tolerated dose (MTD) of MnBuOE. The safety and PK of a multi-dosing protocol was assessed. RESULTS: Peak plasma drug concentration occurred 30 min post-injection. The t 1/2 was defined as 7 h. MnBuOE induced an anaphylactic reaction and prolonged tachycardia. The MTD was defined as 0.25 mg/kg. The dogs were given MTD 3×/week for 2-3 weeks. The highest recorded tissue drug levels were in the lymph nodes (4-6 µM), followed by kidney and liver (2.5, 2.0 uM, respectively). CONCLUSIONS: We obtained critical information regarding the PK and toxicity of MnBuOE in dogs. The acute drug reaction and tachycardia post-injection have not been described in other species and may be specific to canines. The high tissue drug levels in lymph nodes have not been previously reported. MnBuOE accumulation in lymph nodes has important implications for the utility of adjuvant MnBuOE to treat lymphoma. With MnBuOE lymph node accumulation, reduction in the dose and/or administration frequency could be possible, leading to reduced toxicity.


Asunto(s)
Antineoplásicos/administración & dosificación , Riñón/metabolismo , Hígado/metabolismo , Ganglios Linfáticos/metabolismo , Metaloporfirinas/administración & dosificación , Anafilaxia/inducido químicamente , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Enfermedades de los Perros/tratamiento farmacológico , Perros , Semivida , Linfoma/tratamiento farmacológico , Linfoma/veterinaria , Masculino , Dosis Máxima Tolerada , Metaloporfirinas/farmacocinética , Metaloporfirinas/toxicidad , Especificidad de la Especie , Taquicardia/inducido químicamente , Distribución Tisular
7.
Toxicol Appl Pharmacol ; 326: 34-42, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28400118

RESUMEN

Reactive oxygen species are a well-defined therapeutic target for Parkinson's disease (PD) and pharmacological agents that catalytically scavenge reactive species are promising neuroprotective strategies for treatment. Metalloporphyrins are synthetic catalytic antioxidants that mimic the body's own antioxidant enzymes i.e. superoxide dismutases and catalase. The goal of this study was to determine if newly designed metalloporphyrins have enhanced pharmacodynamics including oral bioavailability, longer plasma elimination half-lives, penetrate the blood brain barrier, and show promise for PD treatment. Three metalloporphyrins (AEOL 11216, AEOL 11203 and AEOL 11114) were identified in this study as potential candidates for further pre-clinical development. Each of these compounds demonstrated blood brain barrier permeability by the i.p. route and two of three compounds (AEOL 11203 and AEOL 11114) were orally bioavailable. All of these compounds protected against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity, including dopamine depletion in the striatum, dopaminergic neuronal loss in the substantial nigra, and increased oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain of the mice without inhibiting MPTP metabolism. Daily therapeutic dosing of these metalloporphyrins were well tolerated without accumulation of brain manganese levels or behavioral alterations assessed by open field and rotarod tests. The study identified two orally active metalloporphyrins and one injectable metalloporphyrin as clinical candidates for further development in PD.


Asunto(s)
Antioxidantes/farmacología , Antiparkinsonianos/farmacología , Encéfalo/efectos de los fármacos , Intoxicación por MPTP/prevención & control , Metaloporfirinas/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Administración Oral , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/farmacocinética , Conducta Animal/efectos de los fármacos , Disponibilidad Biológica , Biomarcadores/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Permeabilidad Capilar , Modelos Animales de Enfermedad , Dopamina/metabolismo , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Semivida , Inyecciones Intraperitoneales , Intoxicación por MPTP/etiología , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/fisiopatología , Masculino , Metaloporfirinas/administración & dosificación , Metaloporfirinas/farmacocinética , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacocinética , Prueba de Desempeño de Rotación con Aceleración Constante
8.
Redox Biol ; 12: 864-871, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28454069

RESUMEN

Although radiation therapy can be effective against cancer, potential damage to normal tissues limits the amount that can be safely administered. In central nervous system (CNS), radiation damage to normal tissues is presented, in part, as suppressed hippocampal neurogenesis and impaired cognitive functions. Mn porphyrin (MnP)-based redox active drugs have demonstrated differential effects on cancer and normal tissues in experimental animals that lead to protection of normal tissues and radio- and chemo-sensitization of cancers. To test the efficacy of MnPs in CNS radioprotection, we first examined the tissue levels of three different MnPs - MnTE-2-PyP5+(MnE), MnTnHex-2-PyP5+(MnHex), and MnTnBuOE-2-PyP5+(MnBuOE). Nanomolar concentrations of MnHex and MnBuOE were detected in various brain regions after daily subcutaneous administration, and MnBuOE was well tolerated at a daily dose of 3mg/kg. Administration of MnBuOE for one week before cranial irradiation and continued for one week afterwards supported production and long-term survival of newborn neurons in the hippocampal dentate gyrus. MnP-driven S-glutathionylation in cortex and hippocampus showed differential responses to MnP administration and radiation in these two brain regions. A better understanding of how preserved hippocampal neurogenesis correlates with cognitive functions following cranial irradiation will be helpful in designing better MnP-based radioprotection strategies.


Asunto(s)
Sistema Nervioso Central/química , Irradiación Craneana/efectos adversos , Metaloporfirinas/administración & dosificación , Neurogénesis/efectos de los fármacos , Protectores contra Radiación/administración & dosificación , Animales , Disponibilidad Biológica , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/efectos de la radiación , Corteza Cerebral/química , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/efectos de la radiación , Femenino , Hipocampo/química , Hipocampo/efectos de los fármacos , Hipocampo/efectos de la radiación , Masculino , Metaloporfirinas/farmacocinética , Ratones Endogámicos C57BL , Neurogénesis/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Protectores contra Radiación/farmacocinética
9.
J Inorg Biochem ; 170: 1-7, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28189031

RESUMEN

To develop a water-soluble and tumor-targeted photosensitizer for photodynamic therapy (PDT), a porphyrin framework containing the metal ion gallium(III) was combined with platinum(II)-based groups to produce two new pentacationic metalloporphyrinates, Ga-4cisPtTPyP (5,10,15,20-tetrakis{cis-diammine-chloro-platinum(II)}(4-pyridyl)-porphyrinato gallium(III) hydroxide tetranitrate) and Ga-4transPtTPyP (5,10,15,20-tetrakis{trans-diammine-chloro-platinum(II)} (4-pyridyl)-porphyrinato gallium(III) hydroxide tetranitrate). Both complexes exhibited high singlet oxygen quantum yields (Φ∆) and remarkable photocytotoxicity with appreciable phototoxic indexes (PIs). In particular, Ga-4cisPtTPyP showed a low IC50 value (Colon 26: 0.12µM; Sarcoma 180: 0.08µM) under illumination and its PI up to 1000. With outstanding tumor accumulation (tumor/muscle ratio>9), Ga-4cisPtTPyP almost completely inhibited tumor growth over two weeks in an in vivo PDT assay. These results imply that Ga-4cisPtTPyP could be a promising anticancer agent for use in PDT.


Asunto(s)
Antineoplásicos , Neoplasias del Colon , Metaloporfirinas , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Metaloporfirinas/farmacología , Solubilidad
10.
Int J Toxicol ; 35(4): 438-53, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27098749

RESUMEN

BMX-001, a manganese porphyrin that has anti-inflammatory, antioxidant, and antitumor properties, is being developed as a potential therapeutic for high-grade glioma (HGG) and head and neck (H&N) cancer. An IND has been opened for BMX-001 in the treatment of HGG (NCT02655601) and another is in preparation for H&N. The safety of BMX-001 has been evaluated in a battery of nonclinical Good Laboratory Practice (GLP)-compliant studies. Systemic toxicity has been evaluated using the intended cGMP product administered subcutaneously for periods of up to 5 weeks in both the mouse and the monkey and included toxicokinetic evaluations to characterize systemic exposure and tissue distribution and clearance of BMX-001. In additional GLP studies, BMX-001 was not irritating to the skin or eye and caused no changes in cardiac rate or rhythm or blood pressure. Mixed results for genotoxicity were seen with the weight of evidence indicating that BMX-001 poses no genotoxic risk in humans. In systemic mouse and monkey studies, loading/maintenance dose no observed adverse effect levels were 12/2 mg/kg/dose and 6/2 mg/kg/dose, respectively, with maintenance doses administered every 3 days after the initial loading dose. Systemic data were used to determine a Food and Drug Administration-approved safe starting dose for the initial clinical study in patients with HGG. BMX-001 was detected in analyzed tissues, including the brain, persisting well past the short plasma clearance period. The highest levels of BMX-001 were seen in the liver and kidneys, with amounts in these tissues returning to close to undetectable levels after a 2-week cessation of dosing.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Metaloporfirinas/farmacocinética , Metaloporfirinas/toxicidad , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Encéfalo/metabolismo , Ojo/efectos de los fármacos , Femenino , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Riñón/metabolismo , Hígado/metabolismo , Macaca fascicularis , Masculino , Dosis Máxima Tolerada , Metaloporfirinas/administración & dosificación , Metaloporfirinas/sangre , Ratones , Pruebas de Mutagenicidad , Conejos , Piel/efectos de los fármacos
11.
J Am Chem Soc ; 138(10): 3518-25, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26894555

RESUMEN

The understanding of nanomaterials for targeted cancer therapy is of great importance as physical parameters of nanomaterials have been shown to be strong determinants that can promote cellular responses. However, there have been rare platforms that can vastly tune the core of nanoparticles at a molecular level despite various nanomaterials employed in such studies. Here we show targeted photodynamic therapy (PDT) with Zr(IV)-based porphyrinic metal-organic framework (MOF) nanoparticles. Through a bottom-up approach, the size of MOF nanoparticles was precisely tuned in a broad range with a designed functional motif, built upon selection of building blocks of the MOF. In particular, molecular properties of the porphyrinic linker are maintained in the MOF nanoparticles regardless of their sizes. Therefore, size-dependent cellular uptake and ensuing PDT allowed for screening of the optimal size of MOF nanoparticles for PDT while MOF nanoparticle formulation of the photosensitizer showed better PDT efficacy than that of its small molecule. Additionally, Zr6 clusters in the MOF enabled an active targeting modality through postsynthetic modification, giving even more enhanced PDT efficacy. Together with our finding of size controllability covering a broad range in the nano regime, we envision that MOFs can be a promising nanoplatform by adopting advanced small molecule systems into the tunable framework with room for postsynthetic modification.


Asunto(s)
Nanopartículas del Metal/química , Metaloporfirinas/síntesis química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/síntesis química , Circonio/química , Ácido Fólico/análogos & derivados , Ácido Fólico/síntesis química , Ácido Fólico/farmacocinética , Células HeLa , Humanos , Nanopartículas del Metal/administración & dosificación , Metaloporfirinas/farmacocinética , Terapia Molecular Dirigida , Tamaño de la Partícula , Fármacos Fotosensibilizantes/farmacocinética , Circonio/farmacocinética
12.
Macromol Biosci ; 16(2): 188-97, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26446281

RESUMEN

A mixed micelle system with pH-responsive and photoactive zinc tetra(progaryloxy-phenyl) porphyrin (ZnTPP) core is constructed for encapsulation of doxorubicin (DOX) to facilitate combined chemo-photodynamic therapy in one delivery system. The mixed micelles show low critical micelle concentration, good storage stability, a pH dependent behavior, and better singlet oxygen generation efficiency compared to the star-shaped PEG-ZnTPP micelles, where the aggregation of ZnTPP within the core, resulting in less photoactive effect. The cell viability treated with DOX-loaded mixed micelles shows higher cytotoxicity than single drug loaded micelles under light irradiation. The improved therapeutic efficiency indicates the combined effect of DOX and ZnTPP and this mixed micelle system has potential as dual-modality for cancer treatment.


Asunto(s)
Doxorrubicina , Metaloporfirinas , Micelas , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Células HeLa , Humanos , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Metaloporfirinas/farmacología , Neoplasias/metabolismo , Neoplasias/patología , Oxígeno Singlete/metabolismo
13.
Int J Radiat Oncol Biol Phys ; 93(4): 892-900, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26530759

RESUMEN

PURPOSE: To test the effects of a novel Mn porphyrin oxidative stress modifier, Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin (MnBuOE), for its radioprotective and radiosensitizing properties in normal tissue versus tumor, respectively. METHODS AND MATERIALS: Murine oral mucosa and salivary glands were treated with a range of radiation doses with or without MnBuOE to establish the dose-effect curves for mucositis and xerostomia. Radiation injury was quantified by intravital near-infrared imaging of cathepsin activity, assessment of salivation, and histologic analysis. To evaluate effects of MnBuOE on the tumor radiation response, we administered the drug as an adjuvant to fractionated radiation of FaDu xenografts. Again, a range of radiation therapy (RT) doses was administered to establish the radiation dose-effect curve. The 50% tumor control dose values with or without MnBuOE and dose-modifying factor were determined. RESULTS: MnBuOE protected normal tissue by reducing RT-mediated mucositis, xerostomia, and fibrosis. The dose-modifying factor for protection against xerostomia was 0.77. In contrast, MnBuOE increased tumor local control rates compared with controls. The dose-modifying factor, based on the ratio of 50% tumor control dose values, was 1.3. Immunohistochemistry showed that MnBuOE-treated tumors exhibited a significant influx of M1 tumor-associated macrophages, which provides mechanistic insight into its radiosensitizing effects in tumors. CONCLUSIONS: MnBuOE widens the therapeutic margin by decreasing the dose of radiation required to control tumor, while increasing normal tissue resistance to RT-mediated injury. This is the first study to quantitatively demonstrate the magnitude of a single drug's ability to radioprotect normal tissue while radiosensitizing tumor.


Asunto(s)
Neoplasias de Cabeza y Cuello/radioterapia , Metaloporfirinas/uso terapéutico , Mucosa Bucal/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Glándulas Salivales/efectos de la radiación , Animales , Relación Dosis-Respuesta en la Radiación , Evaluación Preclínica de Medicamentos/métodos , Fibrosis/etiología , Fibrosis/prevención & control , Metaloporfirinas/farmacocinética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Órganos en Riesgo/patología , Órganos en Riesgo/efectos de la radiación , Dosis de Radiación , Protectores contra Radiación/farmacocinética , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Distribución Aleatoria , Glándulas Salivales/patología , Estomatitis/etiología , Estomatitis/prevención & control , Xerostomía/etiología , Xerostomía/prevención & control
14.
PLoS One ; 10(8): e0134779, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26258982

RESUMEN

In ticks, the digestion of blood occurs intracellularly and proteolytic digestion of hemoglobin takes place in a dedicated type of lysosome, the digest vesicle, followed by transfer of the heme moiety of hemoglobin to a specialized organelle that accumulates large heme aggregates, called hemosomes. In the present work, we studied the uptake of fluorescent metalloporphyrins, used as heme analogs, and amitraz, one of the most regularly used acaricides to control cattle tick infestations, by Rhipicephalus (Boophilus) microplus midgut cells. Both compounds were taken up by midgut cells in vitro and accumulated inside the hemosomes. Transport of both molecules was sensitive to cyclosporine A (CsA), a well-known inhibitor of ATP binding cassette (ABC) transporters. Rhodamine 123, a fluorescent probe that is also a recognized ABC substrate, was similarly directed to the hemosome in a CsA-sensitive manner. Using an antibody against conserved domain of PgP-1-type ABC transporter, we were able to immunolocalize PgP-1 in the digest vesicle membranes. Comparison between two R. microplus strains that were resistant and susceptible to amitraz revealed that the resistant strain detoxified both amitraz and Sn-Pp IX more efficiently than the susceptible strain, a process that was also sensitive to CsA. A transcript containing an ABC transporter signature exhibited 2.5-fold increased expression in the amitraz-resistant strain when compared with the susceptible strain. RNAi-induced down-regulation of this ABC transporter led to the accumulation of metalloporphyrin in the digestive vacuole, interrupting heme traffic to the hemosome. This evidence further confirms that this transcript codes for a heme transporter. This is the first report of heme transport in a blood-feeding organism. While the primary physiological function of the hemosome is to detoxify heme and attenuate its toxicity, we suggest that the use of this acaricide detoxification pathway by ticks may represent a new molecular mechanism of resistance to pesticides.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Proteínas de Artrópodos/química , Hemo/química , Intestinos/fisiología , Rhipicephalus/fisiología , Acaricidas/química , Adenosina Trifosfato/química , Animales , Anticuerpos/química , Bovinos , Cromatografía Líquida de Alta Presión , Ciclosporina/química , Femenino , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Unión Proteica , Estructura Terciaria de Proteína , Protoporfirinas/química , Interferencia de ARN , Rodamina 123/química , Rodamina 123/farmacocinética , Infestaciones por Garrapatas/tratamiento farmacológico , Toluidinas/química , Toluidinas/farmacocinética
15.
Int J Mol Sci ; 17(1)2015 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-26729110

RESUMEN

The development of stimulus-responsive photosensitizer delivery systems that carry a high payload of photosensitizers is of great importance in photodynamic therapy. In this study, redox-responsive polysilsesquioxane nanoparticles (PSilQNPs) built by a reverse microemulsion approach using 5,10,15,20-tetrakis(carboxyphenyl) porphyrin (TCPP) silane derivatives as building blocks, were successfully fabricated. The structural properties of TCPP-PSilQNPs were characterized by dynamic light scattering (DLS)/ζ-potential, scanning electron microscopy (SEM) and thermogravimetric analysis (TGA). The photophysical properties were determined by UV-vis and fluorescence spectroscopy. The quantity of singlet oxygen generated in solution was measured using 1,3-diphenylisobenzofuran. The redox-responsive release of TCPP molecules was successfully demonstrated in solution in the presence of a reducing agent. The internalization of TCPP-PSilQNPs in cancer cells was investigated using laser scanning confocal microscopy. Phototoxicity experiments in vitro showed that the redox-responsive TCPP-PSilQNPs exhibited an improved phototherapeutic effect on cervical cancer cells compared to a non-responsive TCPP-PSilQNP control material.


Asunto(s)
Preparaciones de Acción Retardada/química , Metaloporfirinas/administración & dosificación , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Compuestos de Organosilicio/química , Fármacos Fotosensibilizantes/administración & dosificación , Células HeLa , Humanos , Metaloporfirinas/farmacocinética , Metaloporfirinas/uso terapéutico , Neoplasias/metabolismo , Oxidación-Reducción , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/uso terapéutico
16.
Anticancer Res ; 34(8): 4583-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25075103

RESUMEN

BACKGROUND: Recently, 5-aminolevulinic acid (5-ALA), precursors of protoporphyrin IX (PpIX), and Sn(IV) chlorin e6 (SnCe6) have been proposed as possible sonosensitizers for sonodynamic therapy of cancer. Therefore, we evaluated the pharmacokinetic properties and sonosensitizing activities of 5-ALA and SnCe6 in vivo by using the EMT6/KU tumor-bearing chick embryos. RESULTS: The concentration of PpIX in tumor and liver tissues and serum increased in a time-dependent manner after the i.v. administration of 5-ALA; PpIX concentrations reached their peak level after 5-7 h. The concentration of SnCe6 reached its maximum value in the tumor tissue and serum immediately after i.v. administration. The combined treatment of 5-ALA or SnCe6 with ultrasound irradiation showed a significant antitumor effect towards EMT6/KU solid tumors. CONCLUSION: We evaluated the pharmacokinetic properties and sonosensitizing activities of 5-ALA and SnCe6 in a chick embryo model and found that 5-ALA might be more suitable as a sonosensitizer than SnCe6.


Asunto(s)
Ácido Aminolevulínico/farmacología , Metaloporfirinas/farmacología , Neoplasias Experimentales/terapia , Terapia por Ultrasonido , Ácido Aminolevulínico/farmacocinética , Animales , Línea Celular Tumoral , Embrión de Pollo , Metaloporfirinas/farmacocinética , Ratones , Protoporfirinas/metabolismo
17.
Genesis ; 52(4): 309-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24616213

RESUMEN

Photodynamic therapy utilizes light, a photosensitizer, and molecular oxygen as a treatment modality for a variety of cancers. We have recently combined ruthenium(II) polypyridyl groups with a zinc(II) centered porphyrin as a new photosensitizer for the treatment of melanoma. In-vitro studies have indicated that this photosensitizer is toxic to melanoma cells when irradiated with low energy light; however, it is nontoxic to normal cells under similar conditions. To determine the toxicity and cell viability of this compound in-vivo we present, herein, a study using Drosophila melanogaster. In the absence of light, the new photosensitizer shows no discernible effects to fly larvae at various concentrations of compound and stages of larval development. When the larvae were fed the photosensitizer it was observed, by fluorescence microscopy, that the compound passes through the cell membrane and localizes in the cytosol at lower concentrations and the nucleus at slightly higher concentrations indicating that the compound is not immediately metabolized.


Asunto(s)
Metaloporfirinas/toxicidad , Fármacos Fotosensibilizantes/toxicidad , Rutenio/toxicidad , Zinc/toxicidad , Animales , Encéfalo/metabolismo , Drosophila melanogaster , Ensayos de Selección de Medicamentos Antitumorales , Larva/efectos de los fármacos , Larva/metabolismo , Metaloporfirinas/farmacocinética , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacocinética , Rutenio/farmacocinética , Zinc/farmacocinética
18.
J Biol Inorg Chem ; 19(2): 229-35, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24407461

RESUMEN

As the first clinically approved gadolinium-based blood-pool MRI contrast agent, gadofosveset was designed to bind to human serum albumin (HSA) reversibly, extending the circulation time in the bloodstream. This valuable pharmacokinetic property required for vasculature imaging, however, raises the risk of release and accumulation of gadolinium in vivo. The binding of gadofosveset to HSA significantly increases the relaxivity at low field, which decreases drastically when the magnetic field increases, limiting the applications of gadofosveset at fields of 3 T and higher. To address those challenges, we evaluated a novel dimeric manganese(III) porphyrin (MnP2) in vitro and in vivo as a potential gadolinium-free blood-pool agent. Through multiple spectroscopic studies, we demonstrated that MnP2 binds to HSA tightly. MnP2 exhibits a moderate relaxivity decrease on HSA binding. Nevertheless, owing to the unique field-dependent relaxation behaviors and the dimeric construct (two Mn(III) ions per complex), MnP2-HSA has a molar relaxivity twice that of the gadofosveset-HSA complex at 3 T. Through intravenous injection in rats, MnP2 exhibits long retention and significant contrast enhancement in the vascular compartment, as tested in a 3-T high-field clinical MRI scanner. Taken together, these data demonstrate that MnP2 represents a new class of gadolinium-free blood-pool agents suitable for both regular and high-field applications.


Asunto(s)
Dimerización , Imagen por Resonancia Magnética/métodos , Manganeso/química , Metaloporfirinas/metabolismo , Albúmina Sérica/metabolismo , Animales , Medios de Contraste/química , Medios de Contraste/metabolismo , Medios de Contraste/farmacocinética , Femenino , Humanos , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Unión Proteica , Ratas
19.
Antioxid Redox Signal ; 20(15): 2361-71, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-23758052

RESUMEN

AIMS: The goal of this study was to use two manganese (Mn)-based superoxide dismutase (SOD) mimics to test the hypothesis that reactive oxygen species contribute to both acute and long-term outcomes in a galactose-1P uridylyltransferase (GALT)-null Drosophila melanogaster model of classic galactosemia. RESULTS: We tested the impact of each of two Mn porphyrin SOD mimics, MnTnBuOE-2-PyP(5+), and MnTE-2-PyP(5+), (i) on survival of GALT-null Drosophila larvae reared in the presence versus absence of dietary galactose and (ii) on the severity of a long-term movement defect in GALT-null adult flies. Both SOD mimics conferred a significant survival benefit to GALT-null larvae exposed to galactose but not to controls or to GALT-null larvae reared in the absence of galactose. One mimic, MnTE-2-PyP(5+), also largely rescued a galactose-independent long-term movement defect otherwise seen in adult GALT-null flies. The survival benefit of both SOD mimics occurred despite continued accumulation of elevated galactose-1P in the treated animals, and studies of thiolated proteins demonstrated that in both the presence and absence of dietary galactose MnTE-2-PyP(5+) largely prevented the elevated protein oxidative damage otherwise seen in GALT-null animals relative to controls. INNOVATION AND CONCLUSIONS: Our results confirm oxidative stress as a mediator of acute galactose sensitivity in GALT-null Drosophila larvae and demonstrate for the first time that oxidative stress may also contribute to galactose-independent adult outcomes in GALT deficiency. Finally, our results demonstrate for the first time that both MnTnBuOE-2-PyP(5+) and MnTE-2-PyP(5+) are bioavailable and effective when administered through an oral route in a D. melanogaster model of classic galactosemia.


Asunto(s)
Galactosemias/metabolismo , Imitación Molecular , Superóxido Dismutasa/metabolismo , Animales , Cisteína/sangre , Cisteína/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster , Galactosa/metabolismo , Galactosemias/tratamiento farmacológico , Galactosemias/genética , Galactosemias/mortalidad , Glutatión/sangre , Glutatión/metabolismo , Masculino , Redes y Vías Metabólicas , Metaloporfirinas/farmacocinética , Metaloporfirinas/farmacología , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/química , Superóxido Dismutasa/farmacología
20.
J Med Chem ; 57(2): 516-20, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24328058

RESUMEN

Mn(III) porphyrin (MnP) holds the promise of addressing the emerging challenges associated with Gd-based clinical MRI contrast agents (CAs), namely, Gd-related adverse effect and decreasing sensitivity at high clinical magnetic fields. Two complementary strategies for developing new MnPs as Gd-free CAs with optimized biocompatibility were established to improve relaxivity or clearance rate. MnPs with distinct and tunable pharmacokinetic properties can consequently be constructed for different in vivo applications at clinical field of 3 T.


Asunto(s)
Compuestos de Bifenilo/síntesis química , Medios de Contraste/síntesis química , Complejos de Coordinación/síntesis química , Manganeso , Metaloporfirinas/síntesis química , Porfirinas/síntesis química , Animales , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacocinética , Medios de Contraste/química , Medios de Contraste/farmacocinética , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Gadolinio , Imagen por Resonancia Magnética , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Porfirinas/química , Porfirinas/farmacocinética , Ratas , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA