Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 487, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216556

RESUMEN

Periodontal disease is a significant burden for oral health, causing progressive and irreversible damage to the support structure of the tooth. This complex structure, the periodontium, is composed of interconnected soft and mineralised tissues, posing a challenge for regenerative approaches. Materials combining silicon and lithium are widely studied in periodontal regeneration, as they stimulate bone repair via silicic acid release while providing regenerative stimuli through lithium activation of the Wnt/ß-catenin pathway. Yet, existing materials for combined lithium and silicon release have limited control over ion release amounts and kinetics. Porous silicon can provide controlled silicic acid release, inducing osteogenesis to support bone regeneration. Prelithiation, a strategy developed for battery technology, can introduce large, controllable amounts of lithium within porous silicon, but yields a highly reactive material, unsuitable for biomedicine. This work debuts a strategy to lithiate porous silicon nanowires (LipSiNs) which generates a biocompatible and bioresorbable material. LipSiNs incorporate lithium to between 1% and 40% of silicon content, releasing lithium and silicic acid in a tailorable fashion from days to weeks. LipSiNs combine osteogenic, cementogenic and Wnt/ß-catenin stimuli to regenerate bone, cementum and periodontal ligament fibres in a murine periodontal defect.


Asunto(s)
Nanocables , beta Catenina , Animales , Ratones , Silicio/farmacología , Porosidad , Litio/farmacología , Ácido Silícico/farmacología , Cemento Dental
3.
Eur J Pharm Sci ; 188: 106497, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37329925

RESUMEN

Semi-solid extrusion (SSE) 3D printing enables flexible designs and dose sizes to be printed on demand and is a suitable tool for fabricating personalized dosage forms. Controlled Expansion of Supercritical Solution (CESS®) is a particle size reduction technology, and it produces particles of a pure active pharmaceutical ingredient (API) in a dry state, suspendable in the printing ink. In the current study, as a model API of poorly water-soluble drug, nanoformed piroxicam (nanoPRX) prepared by CESS® was accommodated in hydroxypropyl methylcellulose- or hydroxypropyl cellulose-based ink formulations to warrant the printability in SSE 3D printing. Importantly, care must be taken when developing nanoPRX formulations to avoid changes in their polymorphic form or particle size. Printing inks suitable for SSE 3D printing that successfully stabilized the nanoPRX were developed. The inks were printed into films with escalating doses with exceptional accuracy. The original polymorphic form of nanoPRX in the prepared dosage forms was not affected by the manufacturing process. In addition, the conducted stability study showed that the nanoPRX in the prepared dosage form remained stable for at least three months from printing. Overall, the study rationalizes that with nanoparticle-based printing inks, superior dose control for the production of personalized dosage forms of poorly water-soluble drugs at the point-of-care can be achieved.


Asunto(s)
Piroxicam , Impresión Tridimensional , Tecnología , Excipientes , Agua
4.
Nanoscale Res Lett ; 12(1): 74, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28124301

RESUMEN

Silicon-based mesoporous nanoparticles have been extensively studied to meet the challenges in the drug delivery. Functionality of these nanoparticles depends on their properties which are often changing as a function of particle size and surrounding medium. Widely used characterization methods, dynamic light scattering (DLS), and transmission electron microscope (TEM) have both their weaknesses. We hypothesize that conventional light scattering (LS) methods can be used for a rigorous characterization of medium sensitive nanoparticles' properties, like size, stability, and porosity. Two fundamentally different silicon-based nanoparticles were made: porous silicon (PSi) from crystalline silicon and silica nanoparticles (SN) through sol-gel process. We studied the properties of these mesoporous nanoparticles with two different multiangle LS techniques, DLS and static light scattering (SLS), and compared the results to dry-state techniques, TEM, and nitrogen sorption. Comparison of particle radius from TEM and DLS revealed significant overestimation of the DLS result. Regarding to silica nanoparticles, the overestimation was attributed to agglomeration by analyzing radius of gyration and hydrodynamic radius. In case of PSi nanoparticles, strong correlation between LS result and specific surface area was found. Our results suggest that the multiangle LS methods could be used for the size, stability, and structure characterization of mesoporous nanoparticles.

5.
Langmuir ; 32(49): 13020-13029, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27951684

RESUMEN

The effect of adsorption and confinement on ibuprofen was studied by immersion loading the molecules into porous silicon (PSi) microparticles. The PSi microparticles were modified into thermally oxidized PSi (TOPSi) and thermally hydrocarbonized PSi (THCPSi) to evaluate the effects of the loading solvent and the surface chemistry on the obtainable drug payloads. The payloads, location, and the molecular state of the adsorbed drug were evaluated using thermal analysis. The results showed that after the adsorption of ∼800 mg/cm3 (wdrug/vpores) of drug into the mesopores, depending on the solvent used in the immersion, the drug began to rapidly recrystallize on the external surface of the particles. Moderate concentrations, however, enabled payloads of 800-850 mg/cm3 without excessive surface crystallization, and thus, there was no need for rinsing the samples to remove the externally crystallized portion. The results showed that the confined ibuprofen forms nanocrystals inside of the mesopores after approximately 200 mg/cm3 payloads were obtained, accounting for half of the adsorbed drug amount. The presence of both crystalline and noncrystalline phases was further characterized using variable temperature solid-state nuclear magnetic resonance (NMR) measurements. The interactions between the drug molecules and the pore walls of TOPSi and THCPSi were observed using Fourier transform infrared and 1H NMR spectroscopies, and the hydrogen bonding between the silanol groups of TOPSi and the adsorbed ibuprofen was confirmed, but having only limited effect on the overall state of the confined drug. In vitro drug permeation studies in Caco-2 and Caco-2/HT29 cocultures showed that the adsorption onto hydrophilic or hydrophobic PSi microparticles had no significant effects on the ibuprofen permeation, whether the drug was partially nanocrystalline or completely in a liquidlike state.


Asunto(s)
Portadores de Fármacos/química , Ibuprofeno/química , Silicio , Adsorción , Células CACO-2 , Cristalización , Humanos , Nanopartículas , Porosidad
6.
Eur J Pharm Sci ; 88: 257-66, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26993963

RESUMEN

The aim of the present study was to synthesize functionalized (18)F-labeled NLs ((18)F-NLs) and evaluate their biological behavior in mouse models of Alzheimer's disease (AD) using positron emission tomography (PET) and ex vivo brain autoradiography. (18)F-fluorine was introduced to (18)F-NLs either by using a core forming (18)F-lipid or by encapsulating a (18)F-tracer, (18)F-treg-curcumin inside the NLs. Phosphatidic acid (PA) and curcumin derivative (Curc) functionalized (18)F-NLs with or without additional mApoE functionalization were produced using thin film hydration. The biodistribution and ß-amyloid plaque-binding ability of (18)F-NLs were studied in wild type mice and AD mouse models using in vivo PET imaging and ex vivo brain autoradiography at 60min after (18)F-NL injection. Functionalized (18)F-NLs were successfully synthesized. The preclinical evaluation in mice showed that the functional group affected the biodistribution of (18)F-NLs. Further functionalization with mApoE increased the brain-to-blood ratio of (18)F-NLs but the overall brain uptake remained low with all functionalized (18)F-NLs. The liposomal encapsulation of (18)F-treg-curcumin was not successful and preclinical results of encapsulated (18)F-treg-curcumin and plain (18)F-treg-curcumin were identical. Although the studied functionalized (18)F-NLs were not suitable for PET imaging as such, the synthesis techniques introduced in this study can be utilized to modify the biological behavior of (18)F-labeled NLs.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/metabolismo , Flúor/metabolismo , Liposomas/farmacocinética , Nanoestructuras/química , Animales , Autorradiografía/métodos , Femenino , Flúor/química , Radioisótopos de Flúor , Genes Transgénicos Suicidas , Liposomas/química , Masculino , Ratones , Estructura Molecular , Tomografía de Emisión de Positrones , Radiofármacos/síntesis química , Tecnología Farmacéutica , Distribución Tisular
7.
Mol Pharm ; 12(7): 2254-64, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-26035734

RESUMEN

The oral bioavailability of a poorly water-soluble drug is often inadequate for the desired therapeutic effect. The bioavailability can be improved by enhancing the physicochemical properties of the drug (e.g., dissolution rate, permeation across the gastrointestinal tract). Other approach include shielding the drug from the gastric metabolism and targeted drug release to obtain optimal drug absorption. In this study, a poorly water-soluble model drug, griseofulvin, was encapsulated as disordered solid dispersions into Eudragit L 100-55 enteric polymer micromatrix particles, which were produced by electrospraying. Similar micromatrix particles were also produced with griseofulvin-loaded thermally oxidized mesoporous silicon (TOPSi) nanoparticles dispersed to the polymer micromatrices. The in vitro drug dissolution at pH 1.2 and 6.8, and permeation at pH 7.4 across Caco-2/HT29 cell monolayers from the micromatrix particles, were investigated. The micromatrix particles were found to be gastro-resistant, while at pH 6.8 the griseofulvin was released very rapidly in a fast-dissolving form. Compared to free griseofulvin, the permeability of encapsulated griseofulvin across the intestinal cell monolayers was greatly improved, particularly for the TOPSi-doped micromatrix particles. The griseofulvin solid dispersions were stable during storage for 6 months at accelerated conditions. Overall, the method developed here could prove to be a useful oral drug delivery solution for improving the bioavailability of poorly water-soluble or otherwise problematic drugs.


Asunto(s)
Griseofulvina/química , Polímeros/química , Resinas Acrílicas/química , Disponibilidad Biológica , Células CACO-2 , Línea Celular Tumoral , Portadores de Fármacos/química , Griseofulvina/farmacocinética , Células HT29 , Humanos , Absorción Intestinal/efectos de los fármacos , Nanopartículas/química , Permeabilidad , Silicio/química , Solubilidad , Tecnología Farmacéutica/métodos , Agua/química
8.
Acta Biomater ; 16: 206-14, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25637067

RESUMEN

Dual-drug delivery of antiangiogenic and chemotherapeutic drugs can enhance the therapeutic effect for cancer therapy. Conjugation of methotrexate (MTX) to porous silicon (PSi) nanoparticles (MTX-PSi) with positively charged surface can improve the cellular uptake of MTX and inhibit the proliferation of cancer cells. Herein, MTX-PSi conjugates sustained the release of MTX up to 96 h, and the released fragments including MTX were confirmed by mass spectrometry. The intracellular distribution of the MTX-PSi nanoparticles was confirmed by transmission electron microscopy. Compared to pure MTX, the MTX-PSi achieved similar inhibition of cell proliferation in folate receptor (FR) over-expressing U87 MG cancer cells, and a higher effect in low FR-expressing EA.hy926 cells. Nuclear fragmentation analysis demonstrated programmed cell apoptosis of MTX-PSi in the high/low FR-expressing cancer cells, whereas PSi alone at the same dose had a minor effect on cell apoptosis. Finally, the porous structure of MTX-PSi enabled a successful concomitant loading of another anti-angiogenic hydrophobic drug, sorafenib, and considerably enhanced the dissolution rate of sorafenib. Overall, the MTX-PSi nanoparticles can be used as a platform for combination chemotherapy by simultaneously enhancing the dissolution rate of a hydrophobic drug and sustaining the release of a conjugated chemotherapeutic drug.


Asunto(s)
Sistemas de Liberación de Medicamentos , Endocitosis , Nanopartículas/química , Silicio/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Preparaciones de Acción Retardada , Endocitosis/efectos de los fármacos , Humanos , Metotrexato/química , Metotrexato/farmacología , Nanopartículas/ultraestructura , Niacinamida/análogos & derivados , Niacinamida/farmacología , Tamaño de la Partícula , Compuestos de Fenilurea/farmacología , Porosidad , Sorafenib , Espectroscopía Infrarroja por Transformada de Fourier , Electricidad Estática
9.
Langmuir ; 31(5): 1722-9, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25604519

RESUMEN

Porous silicon (PSi) nanoparticles' tunable properties are facilitating their use at highly challenging medical tasks such as peptide delivery. Because of many different mechanisms that are affecting the interaction between the peptide and the particle, the drug incorporation into the mesoporous delivery system is not straightforward. We have studied the adsorption and loading of incretin hormone glucagon like peptide 1 (GLP-1) on PSi nanoparticles. The results show that the highest loading degree can be achieved in pH values near the isoelectric point of peptide, and the phenomenon is independent of the surface's zeta potential. In order to study the interaction between the peptide and the nanoparticle, we studied the adsorption with lower concentrations and noticed that also non-Coulombic forces have a big role in adsorption of GLP-1. Adsorption is effective and pH-independent especially on low peptide concentrations and onto more hydrophobic nanoparticles. Reversibility of adsorption was studied as a function of buffer pH. When the loading is compared to the total mass of the formulation, the loading degree is 29%, and during desorption experiments 25% is released in 4 h and can be considered as a reversible loading degree. Thus, the peptides adsorbed first seem to create irreversibly adsorbed layer that facilitates reversible adsorption of following peptides.


Asunto(s)
Péptido 1 Similar al Glucagón/química , Nanopartículas/química , Silicio/química , Adsorción , Secuencia de Aminoácidos , Péptido 1 Similar al Glucagón/uso terapéutico , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Porosidad , Propiedades de Superficie
10.
Biomaterials ; 35(33): 9224-35, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25123922

RESUMEN

Nanoparticles (NPs) have been suggested for immunotherapy applications in order to optimize the delivery of immuno-stimulative or -suppressive molecules. However, low attention towards the impact of the NPs' physicochemical properties has presented a major hurdle for developing efficient immunotherapeutic agents. Here, the effects of porous silicon (PSi) NPs with different surface chemistries were evaluated on human monocyte-derived dendritic cells (MDDCs) and lymphocytes in order to highlight the importance of the NPs selection in immuno-stimulative or -suppressive treatment. Although all the PSi NPs showed high biocompatibility, only thermally oxidized PSi (TOPSi) and thermally hydrocarbonized PSi (THCPSi) NPs were able to induce very high rate of immunoactivation by enhancing the expression of surface co-stimulatory markers of the MDDCs (CD80, CD83, CD86, and HLA-DR), inducing T-cell proliferation, and also the secretion of interleukins (IL-1ß, IL-4, IL-6, IL-10, IL-12, IFN-γ, and TNF-α). These results indicated a balanced increase in the secretion of Th1, Th2, and Treg cytokines. Moreover, undecylenic acid functionalized THCPSi, as well as poly(methyl vinyl ether-alt-maleic acid) conjugated to (3-aminopropyl)triethoxysilane functionalized thermally carbonized PSi and polyethyleneimine conjugated undecylenic acid functionalized THCPSi NPs showed moderate immunoactivation due to the mild increase in the above-mentioned markers. By contrast, thermally carbonized PSi (TCPSi) and (3-aminopropyl)triethoxysilane functionalized TCPSi NPs did not induce any immunological responses, suggesting that their application could be in the delivery of immunosuppressive molecules. Overall, our findings suggest all the NPs containing more nitrogen or oxygen on the outermost backbone layer have lower immunostimulatory effect than NPs with higher C-H structures on the surface.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Nanopartículas/química , Silicio/química , Silicio/farmacología , Materiales Biocompatibles/química , Proliferación Celular/efectos de los fármacos , Células Dendríticas/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Polietileneimina/química , Porosidad , Propilaminas , Silanos/química , Propiedades de Superficie , Factor de Necrosis Tumoral alfa/metabolismo
11.
Nanoscale ; 6(17): 10377-87, 2014 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-25074521

RESUMEN

Active targeting of nanoparticles to receptor-overexpressing cancer cells has great potential for enhancing the cellular uptake of nanoparticles and for reducing fast clearance of the nanoparticles from the body. Herein, we present a preparation method of a porous silicon (PSi)-based nanodelivery system for breast cancer targeting, by covalently conjugating a synthesized amide-modified hyaluronic acid (HA(+)) derived polymer on the surface of undecylenic acid-modified thermally hydrocarbonized PSi (UnTHCPSi) nanoparticles. The resulting UnTHCPSi-HA(+) nanoparticles showed relatively small size, reduced polydispersibility, high biocompatibility, improved colloidal and human plasma stability, as well as enhanced cellular interactions and internalization. Moreover, we demonstrated that the enhanced cellular association of UnTHCPSi-HA(+) relies on the capability of the conjugated HA(+) to bind and consequently target CD44 receptors expressed on the surface of breast cancer cells, thus making the HA(+)-functionalized UnTHCPSi nanoparticles a suitable and promising nanoplatform for the targeting of CD44-overexpressing breast tumors and for drug delivery.


Asunto(s)
Aminas Biogénicas/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Ácido Hialurónico/química , Nanocápsulas/química , Silicio/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Aminas Biogénicas/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difusión , Composición de Medicamentos/métodos , Humanos , Ensayo de Materiales , Nanocápsulas/administración & dosificación , Nanoconjugados , Nanoporos/ultraestructura , Tamaño de la Partícula , Porosidad , Resultado del Tratamiento
12.
Biomaterials ; 35(26): 7488-500, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24906344

RESUMEN

The development of a stable vehicle with low toxicity, high cellular internalization, efficient endosomal escape, and optimal drug release profile is a key bottleneck in nanomedicine. To overcome all these problems, we have developed a successful layer-by-layer method to covalently conjugate polyethyleneimine (PEI) and poly(methyl vinyl ether-co-maleic acid) (PMVE-MA) copolymer on the surface of undecylenic acid functionalized thermally hydrocarbonized porous silicon nanoparticles (UnTHCPSi NPs), forming a bilayer zwitterionic nanocomposite containing free positive charge groups of hyper-branched PEI disguised by the PMVE-MA polymer. The surface smoothness, charge and hydrophilicity of the developed NPs considerably improved the colloidal and plasma stabilities via enhanced suspensibility and charge repulsion. Furthermore, despite the surface negative charge of the bilayer polymer-conjugated NPs, the cellular trafficking and endosomal escape were significantly increased in both MDA-MB-231 and MCF-7 breast cancer cells. Remarkably, we also showed that the conjugation of surface free amine groups of the highly toxic UnTHCPSi-PEI (Un-P) NPs to the carboxylic groups of PMVE-MA renders acceptable safety features to the system and preserves the endosomal escape properties via proton sponge mechanism of the free available amine groups located inside the hyper-branched PEI layer. Moreover, the double layer protection not only controlled the aggregation of the NPs and reduced the toxicity, but also sustained the drug release of an anticancer drug, methotrexate, with further improved cytotoxicity profile of the drug-loaded particles. These results provide a proof-of-concept evidence that such zwitterionic polymer-based PSi nanocomposites can be extensively used as a promising candidate for cytosolic drug delivery.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/metabolismo , Endosomas/metabolismo , Maleatos/metabolismo , Metotrexato/administración & dosificación , Nanopartículas/metabolismo , Polietileneimina/metabolismo , Polietilenos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Preparaciones de Acción Retardada/química , Femenino , Humanos , Iones/química , Iones/metabolismo , Células MCF-7 , Maleatos/química , Metotrexato/farmacología , Nanopartículas/química , Nanopartículas/ultraestructura , Polietileneimina/química , Polietilenos/química , Polímeros , Porosidad , Silicio , Propiedades de Superficie
13.
Macromol Rapid Commun ; 35(6): 624-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24497275

RESUMEN

Currently, developing a stable nanocarrier with high cellular internalization and low toxicity is a key bottleneck in nanomedicine. Here, we have developed a successful method to covalently conjugate poly(methyl vinyl ether-co-maleic acid) (PMVE-MA) copolymer on the surface of (3-aminopropyl)triethoxysilane-functionalized thermally carbonized porous silicon nanoparticles (APSTCPSi NPs), forming a surface negatively charged nanovehicle with unique properties. This polymer conjugated NPs could modify surface smoothness, charge, and hydrophilicity of the developed NPs, leading to considerable improvement in the colloidal and plasma stabilities via enhanced suspensibility and charge repulsion. Furthermore, despite the surface negative charge of the polymer-conjugated NPs, the cellular internalization was increased in both MDA-MB-231 and MCF-7 breast cancer cells. These results provide a proof-of-concept evidence that such polymer-based PSi nanocomposite can be extensively used as a promising candidate for intracellular drug delivery.


Asunto(s)
Células/citología , Nanopartículas/química , Polímeros/química , Silicio/química , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular , Humanos , Porosidad
14.
ACS Appl Mater Interfaces ; 6(4): 2884-92, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24450851

RESUMEN

Stratified optical filters with distinct spectral features and layered surface chemistry were prepared on silicon substrates with stepwise anodic porosification and thermal carbonization. The use of differing parameters for successive carbonization treatments enabled the production of hydrolytically stable porous silicon-based layered optical structures where the adsorption of water to the lower layer is inhibited. This enables selective shifting of reflectance bands by means of liquid infiltration. The merit of using thermal carbonization for creating layered functionality was demonstrated by comparing the hydrolytic stability resulting from this approach to other surface chemistries available for Si. The functionality of the stratified optical structures was demonstrated under water and ethanol infiltration, and changes in the adsorption properties after 9 months of storage were evaluated. The changes observed in the structure were explained using simulations based on the transfer matrix method and the Bruggeman effective medium approximation. Scanning electron microscopy was used for imaging the morphology of the porous structure. Finally, the adaptability of the method for preparing complex structures was demonstrated by stacking superimposed rugate structures with several reflective bands.

15.
Biomaterials ; 35(4): 1257-66, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24211082

RESUMEN

Porous silicon (PSi) has been demonstrated as a promising drug delivery vector for poorly water-soluble drugs. Here, a simple and efficient method based on copper-free click chemistry was used to introduce targeting moieties to PSi nanoparticles in order to enhance the intracellular uptake and tumor specific targeting hydrophobic drug delivery. Two RGD derivatives (RGDS and iRGD) with azide-terminated groups were conjugated to bicyclononyne-functionalized PSi nanoparticles via copper-free azide-alkyne cycloaddition. The surface functionalization was performed in aqueous solution at 37 °C for 30 min resulting in conjugation efficiencies of 15.2 and 3.4% (molar ratios) and the nanoparticle size increased from 165.6 nm to 179.6 and 188.8 nm for RGDS and iRGD, respectively. The peptides modification enhanced the cell uptake efficiency of PSi nanoparticles in EA.hy926 cells. PSi-RGDS and PSi-iRGD nanoparticles loaded with sorafenib showed a similar trend for the in vitro antiproliferation activity compared to sorafenib dissolved in dimethyl sulfoxide. Furthermore, sorafenib-loaded PSi-RGDS deliver the drug intracellulary efficiently due to the higher surface conjugation ratio, resulting in enhanced in vitro antiproliferation effect. Our results highlight the surface functionalization methodology for PSi nanoparticles applied here as a universal method to introduce functional moieties onto the surface of PSi nanoparticles and demonstrate their potential active targeting properties for anticancer drug delivery.


Asunto(s)
Alquinos/química , Azidas/química , Portadores de Fármacos/química , Nanopartículas/química , Silicio/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Química Clic , Reacción de Cicloadición , Humanos , Neoplasias/tratamiento farmacológico , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Porosidad , Sorafenib
16.
Biomaterials ; 34(36): 9134-41, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24008034

RESUMEN

Tumour targeting nanoparticles (NPs) have demonstrated great potential for enhancing anticancer drug delivery to tumour sites and for reducing the side effects of chemotherapy. However, many nanoparticulate delivery systems still lack efficient tumour accumulation. In this work, we present a porous silicon (PSi) nanovector functionalized with a tumour-homing peptide, which targets the mammary-derived growth inhibitor (MDGI) expressing cancer cells both in vitro and in vivo, thereby enhancing the accumulation of the NPs in the tumours. We demonstrated that the tumour homing peptide (herein designated as CooP) functionalized thermally hydrocarbonized PSi (THCPSi) NPs homed specifically to the subcutaneous MDGI-expressing xenograft tumours. The THCPSi-CooP NPs were stable in human plasma and their uptake by MDGI-expressing cancer cells measured by confocal microscopy and flow cytometry was significantly increased compared to the non-functionalized THCPSi NPs. After intravenous injections into nude mice bearing MDGI-expressing tumours, effective targeting was detected and THCPSi-CooP NPs showed ~9-fold higher accumulation in the tumour site compared to the control THCPSi NPs. Accumulation of both NPs in the vital organs was negligible.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Péptidos/uso terapéutico , Silicio/química , Adsorción , Animales , Proteínas Sanguíneas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Péptidos/farmacología , Porosidad , Espectroscopía Infrarroja por Transformada de Fourier , Distribución Tisular/efectos de los fármacos
17.
ACS Nano ; 7(8): 6884-93, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23889734

RESUMEN

Influenza A viruses (IAVs) cause recurrent epidemics in humans, with serious threat of lethal worldwide pandemics. The occurrence of antiviral-resistant virus strains and the emergence of highly pathogenic influenza viruses have triggered an urgent need to develop new anti-IAV treatments. One compound found to inhibit IAV, and other virus infections, is saliphenylhalamide (SaliPhe). SaliPhe targets host vacuolar-ATPase and inhibits acidification of endosomes, a process needed for productive virus infection. The major obstacle for the further development of SaliPhe as antiviral drug has been its poor solubility. Here, we investigated the possibility to increase SaliPhe solubility by loading the compound in thermally hydrocarbonized porous silicon (THCPSi) nanoparticles. SaliPhe-loaded nanoparticles were further investigated for the ability to inhibit influenza A infection in human retinal pigment epithelium and Madin-Darby canine kidney cells, and we show that upon release from THCPSi, SaliPhe inhibited IAV infection in vitro and reduced the amount of progeny virus in IAV-infected cells. Overall, the PSi-based nanosystem exhibited increased dissolution of the investigated anti-IAV drug SaliPhe and displayed excellent in vitro stability, low cytotoxicity, and remarkable reduction of viral load in the absence of organic solvents. This proof-of-principle study indicates that PSi nanoparticles could be used for efficient delivery of antivirals to infected cells.


Asunto(s)
Amidas/administración & dosificación , Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Nanopartículas/química , Nanotecnología/métodos , Salicilatos/administración & dosificación , Silicio/química , Animales , Perros , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Humanos , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Modelos Químicos , Tamaño de la Partícula , Solventes/química
18.
Biomaterials ; 34(31): 7776-89, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23866976

RESUMEN

Despite steadily increasing insights on the biocompatibility of PSi nanoparticles (NPs), an extensive biosafety study on the immune and red blood cells (RBCs) is still lacking. Herein, we evaluated the impact of the PSi NPs' surface chemistry on immune cells and human RBCs both in vitro and in vivo. Negatively charged hydrophilic and hydrophobic PSi NPs caused less ATP depletion and genotoxicity than the positively charged amine modified hydrophilic PSi NPs, demonstrating the main role of PSi NPs' surface charge on the immunocompatibility profile. Furthermore, cells with lower metabolic activity, longer doubling time, and shorter half-life were more sensitive to the concentration- and time-dependent toxicity in the following order: T-cells ≈ monocytes > macrophages ≈ B-cells. RBC hemolysis and imaging assay revealed a significant correlation between the surface chemistry, the amount of the PSi NPs adsorbed on the cell surface and the extent of morphological changes. The in vivo results showed that despite mild renal steatosis, glomerular degeneration, hepatic central vein dilation and white pulp shrinkage in spleen, no notable changes were observed in the serum level of biochemical and hematological factors. This study is a comprehensive demonstration of the mechanistic direct and indirect genotoxicity effects of PSi NPs, elucidating the most influencing properties for the PSi NPs' design.


Asunto(s)
Eritrocitos/efectos de los fármacos , Nanopartículas/efectos adversos , Nanopartículas/química , Silicio/química , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/ultraestructura , Línea Celular , Eritrocitos/citología , Eritrocitos/ultraestructura , Hemólisis/efectos de los fármacos , Humanos , Masculino , Ratones , Microscopía Electroquímica de Rastreo , Nanopartículas/ultraestructura , Ratas , Ratas Sprague-Dawley
19.
J Control Release ; 170(2): 268-78, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23756152

RESUMEN

Nanoparticulate drug delivery systems offer remarkable opportunities for clinical treatment. However, there are several challenges when they are employed to deliver multiple cargos/payloads, particularly concerning the synchronous delivery of small molecular weight drugs and relatively larger peptides. Since porous silicon (PSi) nanoparticles (NPs) can easily contain high payloads of drugs with various properties, we evaluated their carrier potential in multi-drug delivery for co-loading of the hydrophobic drug indomethacin and the hydrophilic human peptide YY3-36 (PYY3-36). Sequential loading of these two drugs into the PSi NPs enhanced the drug release rate of each drug and also their amount permeated across Caco-2 and Caco-2/HT29 cell monolayers. Regardless of the loading approach used, dual or single, the drug permeation profiles were in good correlation with their drug release behaviour. Furthermore, the permeation studies indicated the critical role of the mucus intestinal layer and the paracellular resistance in the permeation of the therapeutic compounds across the intestinal wall. Loading with PYY3-36 also greatly improved the cytocompatibility of the PSi NPs. Conformational analysis indicated that the PYY3-36 could still display biological activity after release from the PSi NPs and permeation across the intestinal cell monolayers. These results are the first demonstration of the promising potential of PSi NPs for simultaneous multi-drug delivery of both hydrophobic and hydrophilic compounds.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Nanopartículas/administración & dosificación , Silicio/administración & dosificación , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/química , Células CACO-2 , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Portadores de Fármacos/química , Células HT29 , Células Hep G2 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Indometacina/administración & dosificación , Indometacina/química , Ratones , Nanopartículas/química , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Péptido YY/administración & dosificación , Péptido YY/química , Permeabilidad , Porosidad , Silicio/química
20.
Mol Pharm ; 10(1): 353-9, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23186283

RESUMEN

Porous silicon (PSi) is receiving growing attention in biomedical research, for example, in drug and peptide delivery. Inspired by several advantages of PSi, herein, thermally oxidized (TOPSi, hydrophilic), undecylenic acid-treated thermally hydrocarbonized (UnTHCPSi, moderately hydrophilic), and thermally hydrocarbonized (THCPSi, hydrophobic) PSi nanocarriers are investigated for sustained subcutaneous (sc) and intravenous (iv) peptide delivery. The route of administration is shown to affect drastically peptide YY3-36 (PYY3-36) release from the PSi nanocarriers in mice. Subcutaneous nanocarriers are demonstrated to be capable to sustain PYY3-36 delivery over 4 days, with the high absolute bioavailability values of PYY3-36. The pharmacokinetic parameters of PYY3-36 are presented to be similar between the sc PSi nanocarriers despite surface chemistry. In contrast, iv-delivered PSi nanocarriers display significant differences between the surface types. Overall, these results demonstrate the feasibility of PSi nanocarriers for the sustained sc delivery of peptides.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/administración & dosificación , Nanopartículas/química , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Péptido YY/administración & dosificación , Péptido YY/química , Silicio/química , Administración Cutánea , Administración Intravenosa , Animales , Disponibilidad Biológica , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Portadores de Fármacos/administración & dosificación , Sistemas de Liberación de Medicamentos , Ratones , Ratones Endogámicos BALB C , Porosidad , Silicio/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...