Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 19(1): 365, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789274

RESUMEN

BACKGROUND: Tumor phototherapy especially photodynamic therapy (PDT) or photothermal therapy (PTT), has been considered as an attractive strategy to elicit significant immunogenic cell death (ICD) at an optimal tumor retention of PDT/PTT agents. Heptamethine cyanine dye (IR-780), a promising PDT/PTT agent, which can be used for near-infrared (NIR) fluorescence/photoacoustic (PA) imaging guided tumor phototherapy, however, the strong hydrophobicity, short circulation time, and potential toxicity in vivo hinder its biomedical applications. To address this challenge, we developed mesoporous polydopamine nanoparticles (MPDA) with excellent biocompatibility, PTT efficacy, and PA imaging ability, facilitating an efficient loading and protection of hydrophobic IR-780. RESULTS: The IR-780 loaded MPDA (IR-780@MPDA) exhibited high loading capacity of IR-780 (49.7 wt%), good physiological solubility and stability, and reduced toxicity. In vivo NIR fluorescence and PA imaging revealed high tumor accumulation of IR-780@MPDA. Furthermore, the combined PDT/PTT of IR-780@MPDA could induce ICD, triggered immunotherapeutic response to breast tumor by the activation of cytotoxic T cells, resulting in significant suppression of tumor growth in vivo. CONCLUSION: This study demonstrated that the as-developed compact and biocompatible platform could induce combined PDT/PTT and accelerate immune activation via excellent tumor accumulation ability, offering multimodal tumor theranostics with negligible systemic toxicity.


Asunto(s)
Antineoplásicos , Carbocianinas , Colorantes Fluorescentes , Indoles/química , Nanopartículas/química , Polímeros/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Carbocianinas/química , Carbocianinas/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Neoplasias Mamarias Animales , Ratones , Fototerapia , Nanomedicina Teranóstica , Distribución Tisular
2.
Int J Pharm ; 585: 119433, 2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32447023

RESUMEN

Lipid nanocapsules are treasured nanoparticulate systems, although they lack detectability in biological environments. To overcome this, we designed LNCs loaded simultaneously with fluorescent dye and superparamagnetic iron oxide nanoparticles (Dual LNCs). The introduction of both labels did not alter nanoparticle characteristics such as size (50 nm), size distribution (polydispersity index < 0.1) or surface modifications, including the effectiveness of targeting ligands. Furthermore, the colloidal stability, particle integrity and biocompatibility of the nanoparticles were not negatively affected by label incorporation. These Dual LNCs are concomitantly visualizable via fluorescence and transmitted light imaging after either the internalization by cells or systemic administration to mice. Importantly, they are detectable in liver sections of mice using transmission electron microscopy without additional enhancement. The iron content of 0.24% (m/m) is sufficiently high for precise quantification of nanoparticle concentrations via inductively coupled plasma optical emission spectroscopy. Dual LNCs are precious tools for the investigation of in vitro and in vivo performances of lipid nanocapsule formulations, since they allow for the use of complementary imaging methods for broad range detectability.


Asunto(s)
Química Farmacéutica/métodos , Portadores de Fármacos/farmacocinética , Colorantes Fluorescentes/farmacocinética , Lípidos/química , Nanopartículas Magnéticas de Óxido de Hierro/química , Animales , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Células Endoteliales/efectos de los fármacos , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/química , Humanos , Ratones , Ratones de la Cepa 129 , Energía Filtrada en la Transmisión por Microscopía Electrónica , Microscopía Fluorescente , Nanocápsulas/química , Tamaño de la Partícula
3.
Biomater Sci ; 8(9): 2590-2599, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32238997

RESUMEN

In this work we describe the formulation and characterisation of red-emitting polymeric nanocapsules (NCs) incorporating superparamagnetic iron oxide nanoparticles (SPIONs) for magnetic tumour targeting. The self-fluorescent oligomers were synthesised and chemically conjugated to PLGA which was confirmed by NMR spectroscopy, FT-IR spectroscopy and mass spectrometry. Hydrophobic SPIONs were synthesised through thermal decomposition and their magnetic and heating properties were assessed by SQUID magnetometry and calorimetric measurements, respectively. Magnetic nanocapsules (m-NCs) were prepared by a single emulsification/solvent evaporation method. Their in vitro cytotoxicity was examined in CT26 colon cancer cells. The formulated fluorescent m-NCs showed good stability and biocompatibility both in vitro and in vivo in CT 26 colon cancer models. Following intravenous injection, accumulation of m-NCs in tumours was observed by optical imaging. A higher iron content in the tumours exposed to a magnetic field, compared to the contralateral tumours without magnetic exposure in the same animal, further confirmed the magnetic tumour targeting in vivo. The overall results show that the engineered red-emitting m-NCs have great potential as multifunctional nanocarriers for multi-model bioimaging and magnetic-targeted drug delivery.


Asunto(s)
Compuestos Férricos/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Nanocápsulas/administración & dosificación , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Compuestos Férricos/farmacocinética , Colorantes Fluorescentes/farmacocinética , Hipertermia Inducida , Hierro/metabolismo , Fenómenos Magnéticos , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Poliglactina 910/administración & dosificación , Poliglactina 910/farmacocinética , Distribución Tisular
4.
Mol Pharm ; 17(3): 837-851, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31977228

RESUMEN

Delivery efficiencies of theranostic nanoparticles (NPs) based on passive tumor targeting strongly depend either on their blood circulation time or on appropriate modulations of the tumor microenvironment. Therefore, predicting the NP delivery efficiency before and after a tumor microenvironment modulation is highly desirable. Here, we present a new erythrocyte membrane-camouflaged magnetofluorescent nanocarrier (MMFn) with long blood circulation time (92 h) and high delivery efficiency (10% ID for Ehrlich murine tumor model). MMFns owe their magnetic and fluorescent properties to the incorporation of manganese ferrite nanoparticles (MnFe2O4 NPs) and IR-780 (a lipophilic indocyanine fluorescent dye), respectively, to their erythrocyte membrane-derived camouflage. MMFn composition, morphology, and size, as well as optical absorption, zeta potential, and fluorescent, magnetic, and magnetothermal properties, are thoroughly examined in vitro. We then present an analytical pharmacokinetic (PK) model capable of predicting the delivery efficiency (DE) and the time of peak tumor uptake (tmax), as well as changes in DE and tmax due to modulations of the tumor microenvironment, for potentially any nanocarrier. Experimental PK data sets (blood and tumor amounts of MMFns) are simultaneously fit to the model equations using the PK modeling software Monolix. We then validate our model analytical solutions with the numerical solutions provided by Monolix. We also demonstrate how our a priori nonmechanistic model for passive targeting relates to a previously reported mechanistic model for active targeting. All in vivo PK studies, as well as in vivo and ex vivo biodistribution studies, were conducted using two noninvasive techniques, namely, fluorescence molecular tomography (FMT) and alternating current biosusceptometry (ACB). Finally, histopathology corroborates our PK and biodistribution results.


Asunto(s)
Portadores de Fármacos/química , Membrana Eritrocítica/química , Compuestos Férricos/química , Colorantes Fluorescentes/química , Nanopartículas Magnéticas de Óxido de Hierro/química , Imanes/química , Compuestos de Manganeso/química , Terapia Fototérmica/métodos , Animales , Carcinoma de Ehrlich/tratamiento farmacológico , Modelos Animales de Enfermedad , Portadores de Fármacos/farmacocinética , Femenino , Compuestos Férricos/farmacocinética , Colorantes Fluorescentes/farmacocinética , Hipertermia Inducida/métodos , Compuestos de Manganeso/farmacocinética , Ratones , Tamaño de la Partícula , Nanomedicina Teranóstica/métodos , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
5.
Pharm Dev Technol ; 25(3): 366-375, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31835955

RESUMEN

This study aimed at developing an effective in vitro technique for the screening of drug passive diffusion utilising artificial membranes in combination with three selected oils (i.e. cognac, emu, and olive oil). Artificial membranes of varying chemical composition and characteristics have been investigated individually and in combination with the selected oils in terms of the passive diffusion of a fluorescent probe (i.e. Rhodamine 6G or R6G), in a diffusion apparatus as compared to excised pig intestinal tissues. In general, the permeation results showed that the rate and extent of R6G permeation were dependent on the membrane composition as well as the type of oil used. The apparent permeability coefficient (Papp) value for R6G across the cellulose nitrate membrane (0.197 × 10-7 ± 0.069 cm/s) was the closest to the Papp of R6G across the excised pig intestinal tissue (0.210 × 10-7 ± 0.080 cm/s). The cellulose acetate-nitrate mixture membrane impregnated with emu oil also produced a Papp value (0.191 × 10-7 ± 0.010 cm/s) that was relatively close to that of R6G across the excised pig intestinal tissue. The delivery of R6G from gastro-retentive matrix type tablets correlated with the release of R6G from the gastro-retentive tablets.


Asunto(s)
Membranas Artificiales , Aceites/química , Aceites de Plantas/química , Rodaminas/farmacocinética , Animales , Difusión , Colorantes Fluorescentes/farmacocinética , Absorción Intestinal , Aceite de Oliva/química , Permeabilidad , Porcinos , Vitis/química
6.
Eur J Pharm Biopharm ; 139: 186-196, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30951820

RESUMEN

The preclinical evaluation of nasally administered drug candidates requires screening studies based on in vitro models of the nasal mucosa. The aim of this study was to evaluate the morpho-functional characteristics of the 3D MucilAir™ nasal model with a pharmacological focus on [ATP]-binding cassette (ABC) efflux transporters. We initially performed a phenotypic characterization of the MucilAir™ model and assessed its barrier properties by immunofluorescence (IF), protein mass spectrometry and examination of histological sections. We then focused on the functional expression of the ABC transporters P-glycoprotein (P-gp), multidrug resistance associated protein (MRP)1, MRP2 and breast cancer resistance protein (BCRP) in bidirectional transport experiments. The MucilAir™ model comprises a tight, polarized, pseudo-stratified nasal epithelium composed of fully differentiated ciliated, goblet and basal cells. These ABC transporters were all expressed by the cell membranes. P-gp and BCRP were both functional and capable of actively effluxing substrates. The MucilAir™ model could consequently represent a potent tool for evaluating the interaction of nasally administered drugs with ABC transporters.


Asunto(s)
Mucosa Nasal/metabolismo , Técnicas de Cultivo de Tejidos/métodos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Administración Intranasal , Células CACO-2 , Técnicas de Cultivo de Célula , Evaluación Preclínica de Medicamentos/métodos , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/farmacocinética , Voluntarios Sanos , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mucosa Nasal/citología , Proteínas de Neoplasias/metabolismo , Permeabilidad
7.
Bioelectromagnetics ; 39(6): 441-450, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29984850

RESUMEN

Nanosecond electric pulse (nsEP) exposure generates an array of physiological effects. The extent of these effects is impacted by whether the nsEP is a unipolar (UP) or bipolar (BP) exposure. A 600 ns pulse can generate 71% more YO-PRO-1 uptake compared to a 600 ns + 600 ns pulse exposure. This observation is termed "bipolar cancellation" (BPC) because despite the BP nsEP consisting of an additional 600 ns pulse, it generates reduced membrane perturbation. BPC is achieved by varying pulse amplitudes, and symmetrical and asymmetric pulse widths. The effect appears to reverse by increasing the interphase interval between symmetric BP pulses, suggesting membrane recovery is a BPC factor. To date, the impact of the interphase interval between asymmetrical BP and other BPC-inducing symmetrical BP nsEPs has not been fully explored. Additionally, interpulse intervals beyond 50 µs have not been explored to understand the impact of time between the BP nsEP phases. Here, we surveyed different interphase intervals among symmetrical and asymmetrical BP nsEPs to monitor their impact on BPC of YO-PRO-1 uptake. We identified that a 10 microsecond (ms) interphase interval within a symmetrical 600 ns + 600 ns, and 900 ns + 900 ns pulse can resolve BPC. Furthermore, the interphase interval to resolve asymmetric BPC from a 300 ns + 900 ns pulse versus 600 ns pulse exposure is greater (<10 ms) compared to symmetrical BP nsEPs. From these findings, we extended on our conceptual model that BPC is balanced by localized charging and discharging events across the membrane. Bioelectromagnetics. 39:441-450, 2018. Published 2018. This article is a U.S. Government work and is in the public domain in the USA.


Asunto(s)
Membrana Celular/metabolismo , Estimulación Eléctrica/métodos , Animales , Benzoxazoles/farmacocinética , Células CHO , Permeabilidad de la Membrana Celular , Cricetulus , Colorantes Fluorescentes/farmacocinética , Microscopía Confocal , Compuestos de Quinolinio/farmacocinética , Factores de Tiempo
8.
Drug Dev Ind Pharm ; 44(10): 1679-1684, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29939766

RESUMEN

OBJECTIVE: It is difficult to identify the gold nanoparticles (AuNPs) intracellularly due to their non-fluorescent nature. Although gold can quench the fluorescence of any fluorophore, hence it is also difficult to combine gold with a fluorophore such as a semiconductor quantum dots (QDs). The aim of this study was to prepare a single fluorescent stable AuNPs combined with QDs (QDs-Au-NPs) which can be easily detected intracellularly. METHODS: QDs-Au-NPs were prepared via a simple one-step process through controlling the spacing between them using polyethylene glycol (PEG) as space linker in the form of PEGylated QDs. Furthermore, the applicability of this system was evaluated after coating the particles with somatostatin citrate, SST, to active target somatostatin receptors (SSTRs), and identification of the internalized particles via confocal laser scanning spectroscopy. RESULTS: The results showed that the produced Au shell has a thickness of 2.0 ± 0.2 nm and QDs-Au-NPs showed the same fluorescence intensity compared to the unmodified QDs. Additionally, a stable monodisperse QDs-Au-NPs coated with SST were prepared after coating with 11-Mercaptoundecanoic acid. Moreover, cellular uptake study in Human Caucasian breast adenocarcinoma cell lines showed that QDs-Au-SST-NPs could be detected easily using the confocal microscope. In addition, they showed a significant (p ≤ .05) internalization per cell compared to untreated QDs-Au-NPs as detected by flow cytometry. CONCLUSION: It could be concluded that the produced QDs-Au-NPs has a strong fluorescence property like QDs which enable them to be easily detected after cells internalization.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Oro/química , Nanopartículas del Metal/química , Evaluación Preclínica de Medicamentos/métodos , Colorantes Fluorescentes/farmacocinética , Oro/farmacocinética , Humanos , Células MCF-7
9.
Theranostics ; 8(3): 767-784, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29344305

RESUMEN

Fluorescence image-guided surgery combined with intraoperative therapeutic modalities has great potential for intraoperative detection of oncologic targets and eradication of unresectable cancer residues. Therefore, we have developed an activatable theranostic nanoplatform that can be used concurrently for two purposes: (1) tumor delineation with real-time near infrared (NIR) fluorescence signal during surgery, and (2) intraoperative targeted treatment to further eliminate unresected disease sites by non-toxic phototherapy. Methods: The developed nanoplatform is based on a single agent, silicon naphthalocyanine (SiNc), encapsulated in biodegradable PEG-PCL (poly (ethylene glycol)-b-poly(ɛ-caprolactone)) nanoparticles. It is engineered to be non-fluorescent initially via dense SiNc packing within the nanoparticle's hydrophobic core, with NIR fluorescence activation after accumulation at the tumor site. The activatable nanoplatform was evaluated in vitro and in two different murine cancer models, including an ovarian intraperitoneal metastasis-mimicking model. Furthermore, fluorescence image-guided surgery mediated by this nanoplatform was performed on the employed animal models using a Fluobeam® 800 imaging system. Finally, the phototherapeutic efficacy of the developed nanoplatform was demonstrated in vivo. Results: Our in vitro data suggest that the intracellular environment of cancer cells is capable of compromising the integrity of self-assembled nanoparticles and thus causes disruption of the tight dye packing inside the hydrophobic cores and activation of the NIR fluorescence. Animal studies demonstrated accumulation of activatable nanoparticles at the tumor site following systemic administration, as well as release and fluorescence recovery of SiNc from the polymeric carrier. It was also validated that the developed nanoparticles are compatible with the intraoperative imaging system Fluobeam® 800, and nanoparticle-mediated image-guided surgery provides successful resection of cancer tumors. Finally, in vivo studies revealed that combinatorial phototherapy mediated by the nanoparticles could efficiently eradicate chemoresistant ovarian cancer tumors. Conclusion: The revealed properties of the activatable nanoplatform make it highly promising for further application in clinical image-guided surgery and combined phototherapy, facilitating a potential translation to clinical studies.


Asunto(s)
Neoplasias Experimentales/terapia , Fototerapia/métodos , Espectroscopía Infrarroja Corta/métodos , Cirugía Asistida por Computador/métodos , Nanomedicina Teranóstica/métodos , Animales , Femenino , Colorantes Fluorescentes/farmacocinética , Células HEK293 , Humanos , Lactonas/química , Ratones , Ratones Desnudos , Nanopartículas/química , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/cirugía , Polietilenglicoles/química , Porfirinas/farmacocinética
11.
J Control Release ; 260: 154-163, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28601576

RESUMEN

Photoimmunotherapy (PIT) is an emerging low side effect cancer therapy based on a monoclonal antibody (mAb) conjugated with a near-infrared (NIR) phthalocyanine dye IRDye 700DX. IR700 is fluorescent, can be used as an imaging agent, and also is phototoxic. It induces rapid cell death after exposure to NIR light. PIT induces highly selective cancer cell death, while leaving most of tumor blood vessels unharmed, leading to an effect called super-enhanced permeability and retention (SUPR). SUPR significantly improves the effectiveness of the anticancer drug. Currently, the therapeutic effects of PIT are monitored using the IR700 fluorescent signal based on macroscopic fluorescence reflectance imagery. This technique, however, lacks the resolution and depth information to reveal the intratumor heterogeneity of mAb-IR700 distribution. We applied a minimally invasive two-channel fluorescence fiber imaging system by combining the traditional fluorescence imaging microscope with two imaging fiber bundles (~0.85mm). This method monitored mAb-IR700 distribution and therapeutic effects during PIT at different intratumor locations (e.g., tumor surface vs. deep tumor) in situ and in real time simultaneously. This enabled evaluation of the therapeutic effects in vivo and treatment regimens. The average IR700 fluorescence intensity recovery after PIT to the tumor surface is 91.50%, while it is 100.63% in deep tumors. To verify the results, two-photon microscopy combined with a microprism was also used to record the mAb-IR700 distribution and fluorescence intensity of green fluorescent protein (GFP) at different tumor depths during PIT. After PIT treatment, there was significantly higher IR700 fluorescence recovery in deep tumor than in the tumor surface. This phenomenon can be explained by increased vascular permeability immediately after NIR-PIT. Fluorescence intensity of GFP at the tumor surface decreased significantly more compared to that of deep tumor and in controls (no PIT).


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Carbocianinas/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Inmunoterapia , Neoplasias/terapia , Compuestos Organofosforados/administración & dosificación , Fototerapia , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacocinética , Carbocianinas/química , Carbocianinas/farmacocinética , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Proteínas Fluorescentes Verdes , Inmunoglobulina G/inmunología , Rayos Infrarrojos , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacocinética , Panitumumab , Carga Tumoral
12.
Adv Healthc Mater ; 6(16)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504409

RESUMEN

Intratumoral (IT) drug injections reduce systemic toxicity, but delivered volumes and distribution can be inconsistent. To improve IT delivery paradigms, porphyrin-phospholipid (PoP) liposomes are passively loaded with three hydrophilic cargos: sulforhodamine B, a fluorophore; gadolinium-gadopentetic acid, a magnetic resonance (MR) agent; and oxaliplatin, a colorectal cancer chemotherapeutic. Liposome composition is optimized so that cargo is retained in serum and storage, but is released in less than 1 min with exposure to near infrared light. Light-triggered release occurs with PoP-induced photooxidation of unsaturated lipids and all cargos release concurrently. In subcutaneous murine colorectal tumors, drainage of released cargo is delayed when laser treatment occurs 24 h after IT injection, at doses orders of magnitude lower than systemic ones. Delayed light-triggering results in substantial tumor shrinkage relative to controls a week following treatment, although regrowth occurs subsequently. MR imaging reveals that over this time frame, pools of liposomes within the tumor migrate to adjacent regions, possibly leading to altered spatial distribution during triggered drug release. Although further characterization of cargo loading and release is required, this proof-of-principle study suggests that multimodal theranostic IT delivery approaches hold potential to both guide injections and interpret outcomes, in particular when combined with chemo-phototherapy.


Asunto(s)
Antineoplásicos , Medios de Contraste , Colorantes Fluorescentes , Liposomas , Fotoquimioterapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Medios de Contraste/química , Medios de Contraste/farmacocinética , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Liposomas/química , Liposomas/farmacocinética , Liposomas/farmacología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/farmacología , Oxaliplatino , Fosfolípidos/química , Porfirinas/química
13.
Nanomedicine ; 13(3): 955-963, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27884637

RESUMEN

This study represents a novel phototheranostic nanoplatform based on the near-infrared (NIR) heptamethine cyanine dye, IR775, which is capable of concurrent real-time fluorescence imaging and cancer eradication with combinatorial phototherapy. To achieve water solubility and enhance tumor delivery, the hydrophobic IR775 dye was loaded into a biocompatible polymeric nanoparticle with a diameter of ~40nm and slightly negative surface charge (-2.34mV). The nanoparticle-encapsulated hydrophobic IR775 dye (IR775-NP) is characterized by an enhanced fluorescence quantum yield (16%) when compared to the water soluble analogs such as ICG (2.7%) and IR783 (8%). Furthermore, the developed IR-775-NP efficiently generates both heat and reactive oxygen species under NIR light irradiation, eradicating cancer cells in vitro. Finally, animal studies revealed that the IR775-NP accumulates in cancer tumors after systemic administration, efficiently delineates them with NIR fluorescence signal and completely eradicates chemo resistant cancer tissue after a single dose of combinatorial phototherapy.


Asunto(s)
Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/uso terapéutico , Indoles/farmacocinética , Indoles/uso terapéutico , Neoplasias Ováricas/terapia , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Carbocianinas/farmacocinética , Carbocianinas/uso terapéutico , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/análisis , Humanos , Indoles/administración & dosificación , Indoles/análisis , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/análisis , Imagen Óptica/métodos , Neoplasias Ováricas/diagnóstico por imagen , Ovario/diagnóstico por imagen
14.
Biomed Res Int ; 2017: 3273816, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29349071

RESUMEN

Brucine is the active component in traditional Chinese medicine "Ma-Qian-Zi" (Strychnos nux-vomica Linn), with capabilities of analgesic, anti-inflammatory, anti-tumor and so on. It is crucial how to break through the impact of cuticle skin which reduces the penetration of drugs to improve drug transmission rate. The aim of this study is to improve the local drug concentration by using ultrasound. We used fresh porcine skin to study the effects of ultrasound on the transdermal absorption of brucine under the influence of various acoustic parameters, including frequency, amplitude and irradiation time. The transdermal conditions of yellow-green fluorescent nanoparticles and brucine in skin samples were observed by laser confocal microscopy and ultraviolet spectrophotometry. The results show that under ultrasonic conditions, the permeability of the skin to the fluorescent label and brucine (e.g., the depth and concentration of penetration) is increased compared to its passive diffusion permeability. The best ultrasound penetration can make the penetration depth of more than 110 microns, fluorescent nanoparticles and brucine concentration increased to 2-3 times. This work will provide supportive data on how the brucine is better used for transdermal drug delivery (TDD).


Asunto(s)
Antiinflamatorios , Portadores de Fármacos , Nanopartículas , Estricnina/análogos & derivados , Terapia por Ultrasonido/métodos , Administración Cutánea , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/farmacocinética , Nanopartículas/administración & dosificación , Nanopartículas/química , Absorción Cutánea , Estricnina/administración & dosificación , Estricnina/farmacocinética , Strychnos nux-vomica/química , Porcinos
15.
Yakugaku Zasshi ; 136(12): 1613-1621, 2016.
Artículo en Japonés | MEDLINE | ID: mdl-27904095

RESUMEN

This article describes the development of various probes and immunogens for chemical-biological investigations of food flavonoids. We accomplished a large (gram)-scale asymmetric synthesis of a key intermediate, 5-aminopentyl deoxy epigallocatechin-3-gallate (APDOEGCg; 3), an analogue of green tea polyphenol EGCg, in which the key step was cationic cyclization utilizing neighboring group participation of the gallate carbonyl group. The synthetic APDOEGCg (3) was efficiently converted to a fluorescent probe 18 and an immunogen 19 by utilizing the high reactivity of the amine functional group. We confirmed the usefulness of these probes for imaging studies and the generation of antibodies, respectively. We also describe the efficient synthesis of a positron emission tomography (PET) probe [11C]20 by incorporation of 11C into EGCg (1), for which synthetic 4″-Me-EGCg (20) was utilized as an authentic sample. Our synthetic strategy was also applied for the practical synthesis of nobiletin (21), a polymethoxylated flavone from citrus. Synthetic nobiletin was readily converted to various probes by selective demethylation and incorporation of fluorescein, biotin or 11C. These probes should be useful for a range of biological applications. Detailed examination of the mechanisms and further applications are in progress.


Asunto(s)
Catequina/análogos & derivados , Colorantes Fluorescentes/síntesis química , Alimentos , Polifenoles/síntesis química , Catequina/síntesis química , Catequina/química , Catequina/farmacocinética , Ciclización , Flavonas/síntesis química , Flavonas/química , Flavonas/farmacocinética , Flavonoides , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Polifenoles/química , Polifenoles/farmacocinética , Tomografía Computarizada por Tomografía de Emisión de Positrones , , Vacunas Sintéticas/química
16.
Photodiagnosis Photodyn Ther ; 15: 182-90, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27375183

RESUMEN

BACKGROUND AND OBJECTIVES: Plum-blossom needling might enhance transdermal penetration of topically applied drugs by creating vertical channels. The purpose of this study was to evaluate drug delivery assisted by plum-blossom needling comparing with CO2 laser ablative fractional resurfacing (AFR) using 5-aminolevulinic acid (5-ALA), a porphyrin precursor, as a test drug. MATERIALS AND METHODS: Ex vivo porcine skin was treated with plum-blossom needle(HWATO, Suzhou medical supplies factory Co., Ltd. China) or CO2 laser AFR before topical application of 20% 5-ALA(Sigma-Aldrich, Co., USA)cream, placebo cream and no cream. ALA-induced porphyrin fluorescence was measured by fluorescence microscopy at skin depths down to 1800µm. Needling was done by tapping the skin vertically from 5cm high above quickly. AFR was performed with a 10.6µm wavelength prototype CO2 laser, using stacked single pulses of 3 millisecond and 91.6mJ per pulse. Plum-blossom needling after ALA application was also done. Fluorescence intensity on lesion surface was examined by curalux spectrum analyzer (Laser Institute of Munich University, Germany) and VAS pain score was recorded in a randomized split-lesion clinical trial including 6 patients, 8 actinic keratosis lesions. RESULTS: AFR created regular cone-shaped channels surrounded by a 70µm thin layer of thermally coagulated dermis, respectively. The cone is approximately 200µm in diameter at the opening and 1850µm in depth. Plum-blossom needle created irregular cone-shaped channels of approximately 180µm in diameter at the opening and it always drags a tail-which was shaped from the closed deeper channels. There was no porphyrin fluorescence in placebo cream or untreated skin sites. Plum-blossom needling followed by ALA application enhanced drug delivery with significantly higher porphyrin fluorescence at the edge of hole (P<0.005) and 100µm far from the hole (P=0.000) versus AFR followed by ALA application at skin depths of 120 and 500µm. Needling after ALA application presented higher porphyrin fluorescence at the edge of hole at skin depths of 120µm (P<0.005) and lower porphyrin fluorescence at 1000µm deep hole edge, and 100µm far from the hole at 120µm, 500µm and 1000µm depths versus AFR followed by ALA application (P<0.005). Skin massage after ALA application did not affect ALA-induced porphyrin fluorescence after pretreatment of plum-blossom needling or AFR. ALA application after plum-blossom needling was better than before plum-blossom needling. The clinical trial showed that the surface fluorescence intensity was stronger in needle-pretreated-lesion than in laser-pretreated-lesion. While the VAS pain score between needle treatment and laser treatment was almost the same. CONCLUSIONS: Plum-blossom needling facilitates delivery of topical ALA into the dermis. It may help ALA to diffuse a little more broadly than AFR does in superficial dermis and obtain similar clinical effect with a much lower cost. Plum-blossom needling treatment appears to be a clinically practical and economical means for enhancing transdermal delivery of ALA, a photodynamic therapy drug, and presumably many other topical skin medications.


Asunto(s)
Terapia por Acupuntura/métodos , Ácido Aminolevulínico/farmacocinética , Queratosis Actínica/metabolismo , Protoporfirinas/farmacocinética , Absorción Cutánea/fisiología , Piel/metabolismo , Terapia por Acupuntura/instrumentación , Anciano , Animales , Femenino , Colorantes Fluorescentes/farmacocinética , Humanos , Técnicas In Vitro , Queratosis Actínica/patología , Masculino , Microscopía Fluorescente/métodos , Persona de Mediana Edad , Fármacos Fotosensibilizantes/farmacocinética , Piel/patología , Porcinos
17.
Nanomedicine (Lond) ; 11(16): 2059-72, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27465123

RESUMEN

AIM: Formulate nanometric oil droplets for encapsulating solid nanoparticles and assess their interactions with cells. MATERIALS & METHODS: Soybean oil droplets, stabilized by Pluronic F68 surfactant, incorporating hydrophobically modified fluorescent silica, nanoparticles were obtained. Cytotoxicity over time, internalization, subsequent intracellular localization and internalization pathways were assessed by microscopy (fluoresence and TEM) in vitro with HeLa cells. RESULTS: Oil droplets encapsulating solid nanoparticles are readily internalized by HeLa cells like free nanoparticles but the intracellular localization differs (nanoemulsions less colocalized with lysosomes) as well as internalization pathway is used (nanoemulsions partially internalized by nonendocytic transport). No cytotoxicity could be observed for either particles tested. CONCLUSION: Our results confirm that nanometric emulsions encapsulating solid nanoparticles can be used for alternative and multifunctional intracellular delivery.


Asunto(s)
Portadores de Fármacos/química , Emulsiones/química , Colorantes Fluorescentes/administración & dosificación , Nanopartículas/administración & dosificación , Dióxido de Silicio/administración & dosificación , Aceite de Soja/química , Colorantes Fluorescentes/farmacocinética , Células HeLa , Humanos , Microscopía Confocal , Nanopartículas/química , Nanopartículas/ultraestructura , Tamaño de la Partícula , Poloxámero/química , Dióxido de Silicio/farmacocinética , Aceite de Soja/farmacocinética
18.
ACS Appl Mater Interfaces ; 8(28): 17859-69, 2016 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-27351208

RESUMEN

As a novel fluorescent probe in the second near-infrared window, Ag2Se quantum dots (QDs) exhibit great prospect in in vivo imaging due to their maximal penetration depth and negligible background. However, the in vivo behavior and toxicity of Ag2Se QDs still largely remain unknown, which severely hinders their wide-ranging biomedical applications. Herein, we systematically studied the blood clearance, distribution, transformation, excretion, and toxicity of polyethylene glycol (PEG) coated Ag2Se QDs in mice after intravenous administration with a high dose of 8 µmol/kg body weight. QDs are quickly cleared from the blood with a circulation half-life of 0.4 h. QDs mainly accumulate in liver and spleen and are remarkably transformed into Ag and Se within 1 week. Ag is excreted from the body readily through both feces and urine, whereas Se is excreted hardly. The toxicological evaluations demonstrate that there is no overt acute toxicity of Ag2Se QDs to mice. Moreover, in regard to the in vivo stability problem of Ag2Se QDs, the biotransformation and its related metabolism are intensively discussed, and some promising coating means for Ag2Se QDs to avert transformation are proposed as well. Our work lays a solid foundation for safe applications of Ag2Se QDs in bioimaging in the future.


Asunto(s)
Puntos Cuánticos/metabolismo , Puntos Cuánticos/toxicidad , Compuestos de Selenio/farmacocinética , Compuestos de Selenio/toxicidad , Compuestos de Plata/farmacocinética , Compuestos de Plata/toxicidad , Animales , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/toxicidad , Rayos Infrarrojos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/toxicidad , Puntos Cuánticos/química , Distribución Aleatoria , Compuestos de Selenio/sangre , Compuestos de Selenio/química , Compuestos de Plata/sangre , Compuestos de Plata/química , Distribución Tisular
19.
PLoS One ; 10(8): e0136382, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26308617

RESUMEN

Herein, we present a novel imaging platform to study the biological effects of non-invasive radiofrequency (RF) electric field cancer hyperthermia. This system allows for real-time in vivo intravital microscopy (IVM) imaging of radiofrequency-induced biological alterations such as changes in vessel structure and drug perfusion. Our results indicate that the IVM system is able to handle exposure to high-power electric-fields without inducing significant hardware damage or imaging artifacts. Furthermore, short durations of low-power (< 200 W) radiofrequency exposure increased transport and perfusion of fluorescent tracers into the tumors at temperatures below 41°C. Vessel deformations and blood coagulation were seen for tumor temperatures around 44°C. These results highlight the use of our integrated IVM-RF imaging platform as a powerful new tool to visualize the dynamics and interplay between radiofrequency energy and biological tissues, organs, and tumors.


Asunto(s)
Diagnóstico por Imagen , Hipertermia Inducida , Microscopía Intravital/métodos , Neoplasias Mamarias Animales/patología , Ondas de Radio , Algoritmos , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes/farmacocinética , Neoplasias Mamarias Animales/terapia , Ratones , Distribución Tisular
20.
J Vis Exp ; (87)2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24837630

RESUMEN

We describe a multi-angle rotational optical imaging (MAROI) system for in vivo monitoring of physiopathological processes labeled with a fluorescent marker. Mouse models (brain tumor and arthritis) were used to evaluate the usefulness of this method. Saposin C (SapC)-dioleoylphosphatidylserine (DOPS) nanovesicles tagged with CellVue Maroon (CVM) fluorophore were administered intravenously. Animals were then placed in the rotational holder (MARS) of the in vivo imaging system. Images were acquired in 10° steps over 380°. A rectangular region of interest (ROI) was placed across the full image width at the model disease site. Within the ROI, and for every image, mean fluorescence intensity was computed after background subtraction. In the mouse models studied, the labeled nanovesicles were taken up in both the orthotopic and transgenic brain tumors, and in the arthritic sites (toes and ankles). Curve analysis of the multi angle image ROIs determined the angle with the highest signal. Thus, the optimal angle for imaging each disease site was characterized. The MAROI method applied to imaging of fluorescent compounds is a noninvasive, economical, and precise tool for in vivo quantitative analysis of the disease states in the described mouse models.


Asunto(s)
Artritis/diagnóstico , Neoplasias Encefálicas/diagnóstico , Colorantes Fluorescentes/administración & dosificación , Nanoestructuras/administración & dosificación , Óptica y Fotónica/métodos , Fosfatidilserinas/administración & dosificación , Saposinas/administración & dosificación , Absorción , Animales , Artritis/metabolismo , Artritis/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Femenino , Colorantes Fluorescentes/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones Transgénicos , Imagen Óptica , Óptica y Fotónica/instrumentación , Imagen de Cuerpo Entero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA