Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 34(42): e2203915, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35985348

RESUMO

As a result of the deficient tumor-specific antigens, potential off-target effect, and influence of protein corona, metal-organic framework nanoparticles have inadequate accumulation in tumor tissues, limiting their therapeutic effects. In this work, a pH-responsive linker (L) is prepared by covalently modifying oleylamine (OA) with 3-(bromomethyl)-4-methyl-2,5-furandione (MMfu) and poly(ethylene glycol) (PEG). Then, the L is embedded into a solid lipid nanoshell to coat apilimod (Ap)-loaded zeolitic imidazolate framework (Ap-ZIF) to form Ap-ZIF@SLN#L. Under the tumor microenvironment, the hydrophilic PEG and MMfu are removed, exposing the hydrophobic OA on Ap-ZIF@SLN#L, increasing their uptake in cancer cells and accumulation in the tumor. The ZIF@SLN#L nanoparticle induces reactive oxygen species (ROS). Ap released from Ap-ZIF@SLN#L significantly promotes intracellular ROS and lactate dehydrogenase generation. Ap-ZIF@SLN#L inhibits tumor growth, increases the survival rate in mice, activates the tumor microenvironment, and improves the infiltration of macrophages and T cells in the tumor, as demonstrated in two different tumor-bearing mice after injections with Ap-ZIF@SLN#TL. Furthermore, mice show normal tissue structure of the main organs and the normal serum level in alanine aminotransferase and aspartate aminotransferase after treatment with the nanoparticles. Overall, this pH-responsive targeting strategy improves nanoparticle accumulation in tumors with enhanced therapeutic effects.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Coroa de Proteína , Zeolitas , Camundongos , Animais , Estruturas Metalorgânicas/química , Espécies Reativas de Oxigênio , Alanina Transaminase , Anidridos Maleicos , Nanopartículas/química , Zeolitas/química , Neoplasias/tratamento farmacológico , Polietilenoglicóis/química , Concentração de Íons de Hidrogênio , Aspartato Aminotransferases , Lactato Desidrogenases , Lipídeos , Microambiente Tumoral
2.
Int J Pharm ; 624: 121959, 2022 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-35792422

RESUMO

Many drugs with anticancer potential fail in their translation to the clinics due to problems related to pharmacokinetics. LEM2 is a new dual inhibitor of MDM2/mutp53-TAp73 interactions with interesting in vitro anticancer activity, which opens new hopes as an unconventional anticancer therapeutic strategy against cancers lacking p53 or with impaired p53 pathways. As others xanthone derivatives, LEM2 has limited aqueous solubility, posing problems to pursue in vivo assays, and therefore limiting its potential clinical translation. In this work, a mesoporous silicon (PSi)-based nanodelivery system was developed with folate functionalization (APTES-TCPSi-PEG-FA) for targeted delivery, which successfully increased LEM2 solubility when compared to bulk LEM2, evidenced in payload release study. Such effect was reflected on the increase of LEM2 cytotoxicity in HCT116 and MDA-MB-231 cancer cells when treated with LEM2-loaded APTES-TCPSi-PEG-FA, by reducing cell viability lower than 50% in comparison with bulk LEM2. Despite the reduced LEM2 loading degree, which still limits its application in further in vivo assays, the results obtained herein recognize PSi-based nanodelivery systems as a promising strategy to improve LEM2 anticancer activity and bioavailability, which will be relevant for the potential use of this potent TAp73 activator in anticancer therapy.


Assuntos
Antineoplásicos , Nanopartículas , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Ácido Fólico , Silício , Dióxido de Silício , Proteína Supressora de Tumor p53
3.
Adv Mater ; 34(9): e2108012, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34877724

RESUMO

An alternative strategy of choosing photothermal and weak-immunostimulatory porous silicon@Au nanocomposites as particulate cores to prepare a biomimetic nanovaccine is reported to improve its biosafety and immunotherapeutic efficacy for solid tumors. A quantitative analysis method is used to calculate the loading amount of cancer cell membranes onto porous silicon@Au nanocomposites. Assisted with foreign-body responses, these exogenous nanoparticulate cores with weak immunostimulatory effect can still efficiently deliver cancer cell membranes into dendritic cells to activate them and the downstream antitumor immunity, resulting in no occurrence of solid tumors and the survival of all immunized mice during 55 day observation. In addition, this nanovaccine, as a photothermal therapeutic agent, synergized with additional immunotherapies can significantly inhibit the growth and metastasis of established solid tumors, via the initiation of the antitumor immune responses in the body and the reversion of their immunosuppressive microenvironments. Considering the versatile surface engineering of porous silicon nanoparticles, the strategy developed here is beneficial to construct multifunctional nanovaccines with better biosafety and more diagnosis or therapeutic modalities against the occurrence, recurrence, or metastasis of solid tumors in future clinical practice.


Assuntos
Nanocompostos , Nanopartículas , Neoplasias , Animais , Biomimética/métodos , Imunoterapia , Camundongos , Nanopartículas/uso terapêutico , Neoplasias/terapia , Microambiente Tumoral
4.
Cell Rep ; 35(8): 109131, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34038723

RESUMO

Communication between biological components is critical for homeostasis maintenance among the convergence of complicated bio-signals. For therapeutic nanoparticles (NPs), the general lack of effective communication mechanisms with the external cellular environment causes loss of homeostasis, resulting in deprived autonomy, severe macrophage-mediated clearance, and limited tumor accumulation. Here, we develop a multistage signal-interactive system on porous silicon particles through integrating the Self-peptide and Tyr-Ile-Gly-Ser-Arg (YIGSR) peptide into a hierarchical chimeric signaling interface with "don't eat me" and "eat me" signals. This biochemical transceiver can act as both the signal receiver for amantadine to achieve NP transformation and signal conversion as well as the signal source to present different signals sequentially by reversible self-mimicking. Compared with the non-interactive controls, these signal-interactive NPs loaded with AS1411 and tanespimycin (17-AAG) as anticancer drugs improve tumor targeting 2.8-fold and tumor suppression 6.5-fold and showed only 51% accumulation in the liver with restricted hepatic injury.


Assuntos
Comunicação Celular/imunologia , Nanopartículas/metabolismo , Neoplasias/imunologia , Humanos , Modelos Moleculares , Estadiamento de Neoplasias , Transdução de Sinais
5.
Adv Healthc Mater ; 9(17): e2000529, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32729247

RESUMO

Biohybrid nanosystems represent the cutting-edge research in biofunctionalization of micro- and nano-systems. Their physicochemical properties bring along advantages in the circulation time, camouflaging from the phagocytes, and novel antigens. This is partially a result of the qualitative differences in the protein corona, and the preferential targeting and uptake in homologous cells. However, the effect of the cell membrane on the cellular endocytosis mechanisms and time has not been fully evaluated yet. Here, the effect is assessed by quantitative flow cytometry analysis on the endocytosis of hydrophilic, negatively charged porous silicon nanoparticles and on their membrane-coated counterparts, in the presence of chemical inhibitors of different uptake pathways. Principal component analysis is used to analyze all the data and extrapolate patterns to highlight the cell-specific differences in the endocytosis mechanisms. Furthermore, the differences in the composition of static protein corona between naked and coated particles are investigated together with how these differences affect the interaction with human macrophages. Overall, the presence of the cell membrane only influences the speed and the entity of nanoparticles association with the cells, while there is no direct effect on the endocytosis pathways, composition of protein corona, or any reduction in macrophage-mediated uptake.


Assuntos
Nanopartículas , Coroa de Proteína , Membrana Celular , Endocitose , Humanos , Porosidade , Silício
6.
ACS Appl Mater Interfaces ; 12(6): 6899-6909, 2020 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-31967771

RESUMO

Heart tissue engineering is critical in the treatment of myocardial infarction, which may benefit from drug-releasing smart materials. In this study, we load a small molecule (3i-1000) in new biodegradable and conductive patches for application in infarcted myocardium. The composite patches consist of a biocompatible elastomer, poly(glycerol sebacate) (PGS), coupled with collagen type I, used to promote cell attachment. In addition, polypyrrole is incorporated because of its electrical conductivity and to induce cell signaling. Results from the in vitro experiments indicate a high density of cardiac myoblast cells attached on the patches, which stay viable for at least 1 month. The degradation of the patches does not show any cytotoxic effect, while 3i-1000 delivery induces cell proliferation. Conductive patches show high blood wettability and drug release, correlating with the rate of degradation of the PGS matrix. Together with the electrical conductivity and elongation characteristics, the developed biomaterial fits the mechanical, conductive, and biological demands required for cardiac treatment.


Assuntos
Decanoatos/química , Sistemas de Liberação de Medicamentos/métodos , Glicerol/análogos & derivados , Infarto do Miocárdio/tratamento farmacológico , Polímeros/química , Bibliotecas de Moléculas Pequenas/química , Animais , Sistemas de Liberação de Medicamentos/instrumentação , Condutividade Elétrica , Glicerol/química , Humanos , Teste de Materiais , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Nanopartículas/química , Pirróis/química , Bibliotecas de Moléculas Pequenas/farmacologia
7.
RSC Adv ; 10(58): 35198-35205, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-35515680

RESUMO

Erythrocyte-based drug delivery systems have been investigated for their biocompatibility, long circulation time, and capability to transport cargo all around the body, thus presenting enormous potential in medical applications. In this study, we investigated hybrid nanoparticles consisting of nano-sized autologous or allogeneic red blood cell (RBC) membranes encapsulating porous silicon nanoparticles (PSi NPs). These NPs were functionalized with a model cancer antigen TRP2, which was either expressed on the surface of the RBCs by a cell membrane-mimicking block copolymer polydimethylsiloxane-b-poly-2-methyl-2-oxazoline, or attached on the PSi NPs, thus hidden within the encapsulation. When in the presence of peripheral blood immune cells, these NPs resulted in apoptotic cell death of T cells, where the NPs having TRP2 within the encapsulation led to a stronger T cell deletion. The deletion of the T cells did not change the relative proportion of CD4+ and cytotoxic CD8+ T cells. Overall, this work shows the combination of nano-sized RBCs, PSi, and antigenic peptides may have use in the treatment of autoimmune diseases.

8.
Biomaterials ; 227: 119556, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31670035

RESUMO

Generation of new neurons by utilizing the regenerative potential of adult neural stem cells (NSCs) and neuroblasts is an emerging therapeutic strategy to treat various neurodegenerative diseases, including neuronal loss after stroke. Committed to neuronal lineages, neuroblasts are differentiated from NSCs and have a lower proliferation rate. In stroke the proliferation of the neuroblasts in the neurogenic areas is increased, but the limiting factor for regeneration is the poor survival of migrating neuroblasts. Survival of neuroblasts can be promoted by small molecules; however, new drug delivery methods are needed to specifically target these cells. Herein, to achieve specific targeting, we have engineered biofunctionalized porous silicon nanoparticles (PSi NPs) conjugated with a specific antibody against polysialylated neural cell adhesion molecule (PSA-NCAM). The PSi NPs loaded with a small molecule drug, SC-79, were able to increase the activity of the Akt signaling pathway in doublecortin positive neuroblasts both in cultured cells and in vivo in the rat brain. This study opens up new possibilities to target drug effects to migrating neuroblasts and facilitate differentiation, maturation and survival of developing neurons. The conjugated PSi NPs are a novel tool for future studies to develop new therapeutic strategies aiming at regenerating functional neurocircuitry after stoke.


Assuntos
Nanopartículas , Células-Tronco Neurais , Acidente Vascular Cerebral , Animais , Proteína Duplacortina , Porosidade , Ratos , Silício , Acidente Vascular Cerebral/tratamento farmacológico
9.
ACS Nano ; 13(6): 6477-6490, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31100004

RESUMO

Recent approaches in the treatment of cancer focus on involving the immune system to control the tumor growth. The administration of immunotherapies, like checkpoint inhibitors, has shown impressive results in the long term survival of patients. Cancer vaccines are being investigated as further tools to prime tumor-specific immunity. Biomaterials show potential as adjuvants in the formulation of vaccines, and biomimetic elements derived from the membrane of tumor cells may widen the range of antigens contained in the vaccine. Here, we show how mice presenting an aggressive melanoma tumor model treated twice with the complete nanovaccine formulation showed control on the tumor progression, while in a less aggressive model, the animals showed remission and control on the tumor progression, with a modification in the immunological profile of the tumor microenvironment. We also prove that co-administration of the nanovaccine together with a checkpoint inhibitor increases the efficacy of the treatment (87.5% of the animals responding, with 2 remissions) compared to the checkpoint inhibitor alone in the B16.OVA model. Our platform thereby shows potential applications as a cancer nanovaccine in combination with the standard clinical care treatment for melanoma cancers.


Assuntos
Vacinas Anticâncer/imunologia , Melanoma/terapia , Nanopartículas/química , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Membrana Celular/química , Camundongos , Camundongos Endogâmicos C57BL , Microfluídica/métodos , Microambiente Tumoral/imunologia
10.
Contrast Media Mol Imaging ; 2019: 3728563, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30992696

RESUMO

Mesoporous silicon (PSi) is biocompatible and tailorable material with high potential in drug delivery applications. Here, we report of an evaluation of PSi as a carrier platform for theranostics by delivering a radioactive ion beam- (RIB-) based radioactive lanthanoid into tumors in a mouse model of prostate carcinoma. Thermally hydrocarbonized porous silicon (THCPSi) wafers were implanted with 159Dy at the facility for radioactive ion beams ISOLDE located at CERN, and the resulting [159Dy]THCPSi was postprocessed into particles. The particles were intratumorally injected into mice bearing prostate cancer xenografts. The stability of the particles was studied in vivo, followed by ex vivo biodistribution and autoradiographic studies. We showed that the process of producing radionuclide-implanted PSi particles is feasible and that the [159Dy]THCPSi particles stay stable and local inside the tumor over seven days. Upon release of 159Dy from the particles, the main site of accumulation is in the skeleton, which is in agreement with previous studies on the biodistribution of dysprosium. We conclude that THCPSi particles are a suitable platform together with RIB-based radiolanthanoids for theranostic purposes as they are retained after administration inside the tumor and the radiolanthanoid remains embedded in the THCPSi.


Assuntos
Radioisótopos/química , Silício/química , Porosidade , Radioisótopos/análise
11.
Proc Natl Acad Sci U S A ; 116(16): 7744-7749, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30926671

RESUMO

Effective cancer therapies often demand delivery of combinations of drugs to inhibit multidrug resistance through synergism, and the development of multifunctional nanovehicles with enhanced drug loading and delivery efficiency for combination therapy is currently a major challenge in nanotechnology. However, such combinations are more challenging to administer than single drugs and can require multipronged approaches to delivery. In addition to being stable and biodegradable, vehicles for such therapies must be compatible with both hydrophobic and hydrophilic drugs, and release drugs at sustained therapeutic levels. Here, we report synthesis of porous silicon nanoparticles conjugated with gold nanorods [composite nanoparticles (cNPs)] and encapsulate them within a hybrid polymersome using double-emulsion templates on a microfluidic chip to create a versatile nanovehicle. This nanovehicle has high loading capacities for both hydrophobic and hydrophilic drugs, and improves drug delivery efficiency by accumulating at the tumor after i.v. injection in mice. Importantly, a triple-drug combination suppresses breast tumors by 94% and 87% at total dosages of 5 and 2.5 mg/kg, respectively, through synergy. Moreover, the cNPs retain their photothermal properties, which can be used to significantly inhibit multidrug resistance upon near-infrared laser irradiation. Overall, this work shows that our nanovehicle has great potential as a drug codelivery nanoplatform for effective combination therapy that is adaptable to other cancer types and to molecular targets associated with disease progression.


Assuntos
Antineoplásicos , Sistemas de Liberação de Medicamentos/métodos , Nanotubos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/efeitos da radiação , Antineoplásicos/uso terapêutico , Feminino , Ouro , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Nus , Técnicas Analíticas Microfluídicas , Nanomedicina , Nanotubos/química , Nanotubos/efeitos da radiação , Neoplasias Experimentais/tratamento farmacológico , Processos Fotoquímicos , Porosidade , Silício
12.
Talanta ; 196: 277-283, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-30683364

RESUMO

Two automatic methodologies were developed to perform the loading and release assays of drugs from porous silicon (PSi) nanoparticles. 5-Fluorouracil (5-FU) was selected as a model drug, and different functionalized PSi nanoparticles were used. While for drug loading studies the reproducible characteristics of flow systems were explored regarding mixture and volumes taken, in the drug release flow methodology versatility in accommodating different devices around the valve were tested. Fluorescent properties of 5-FU were used with detection limit of 9 × 10-4 mg L-1 and a linear response up to 5 mg L-1 The drug loading and release procedures were optimized in sequential injection analysis (SIA) systems obtaining a huge economy regarding the time spent (4 min towards 2 h). Batch and SIA methods were tested and compared for the different behaviours of the PSi nanoparticles towards both methodologies and no noteworthy differences were obtained with Student's t-test for loading with a calculated t value of 2.04 showing the absence of statistical differences. All the results present a RSD less than 10%. So, the developed automatic methodologies are a viable alternative to load drugs and to study the release profiles from PSi nanoparticles.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Fluoruracila/química , Nanopartículas/química , Silício/química , Tecnologia Farmacêutica/métodos , Liberação Controlada de Fármacos , Porosidade , Propriedades de Superfície
13.
Small ; 15(1): e1804332, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30488562

RESUMO

Nanotechnology employs multifunctional engineered materials in the nanoscale range that provides many opportunities for translational stem cell research and therapy. Here, a cell-penetrating peptide (virus-1 transactivator of transcription)-conjugated, porous silicon nanoparticle (TPSi NP) loaded with the Wnt3a protein to increase both the cell survival rate and the delivery precision of stem cell transplantation via a combinational theranostic strategy is presented. The TPSi NP with a pore size of 10.7 nm and inorganic framework enables high-efficiency loading of Wnt3a, prolongs Wnt3a release, and increases antioxidative stress activity in the labeled mesenchymal stem cells (MSCs), which are highly beneficial properties for cell protection in stem cell therapy for myocardial infarction. It is confirmed that the intracellular aggregation of TPSi NPs can highly amplify the acoustic scattering of the labeled MSCs, resulting in a 2.3-fold increase in the ultrasound (US) signal compared with that of unlabeled MSCs. The translational potential of the designed nanoagent for real-time US imaging-guided stem cell transplantation is confirmed via intramyocardial injection of labeled MSCs in a nude mouse model. It is proposed that the intracellular aggregation of protein drug-loaded TPSi NPs could be a simple but robust strategy for improving the therapeutic effect of stem cell therapy.


Assuntos
Citoproteção , Endocitose , Imageamento Tridimensional , Células-Tronco Mesenquimais/citologia , Nanopartículas/química , Silício/química , Ultrassom , Proteínas Virais/metabolismo , Animais , Antioxidantes/farmacologia , Diferenciação Celular , Sobrevivência Celular , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Camundongos Nus , Miocárdio/metabolismo , Nanopartículas/ultraestrutura , Porosidade , Proteína Wnt3A/metabolismo
14.
Biomaterials ; 185: 322-332, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30267958

RESUMO

Orally administrable drug delivery vehicles are developed to manage incurable inflammatory bowel disease (IBD), however, their therapeutic outcomes are compromised by the side effects of systemic drug exposure. Herein, we use hyaluronic acid functionalized porous silicon nanoparticle to bridge enzyme-responsive hydrogel and pH-responsive polymer, generating a hierarchical structured (nano-in-nano-in-micro) vehicle with programmed properties to fully and sequentially overcome the multiple obstacles for efficiently delivering drugs locally to inflamed sites of intestine. After oral administration, the pH-responsive matrix protects the embedded hybrid nanoparticles containing drug loaded hydrogels against the spatially variable physiological environments of the gastrointestinal tract until they reach the inflamed sites of intestine, preventing premature drug release. The negatively charged hybrid nanoparticles selectively target the inflamed sites of intestine, and gradually release drug in response to the microenvironment of inflamed intestine. Overall, the developed hierarchical structured and programmed vehicles load, protect, transport and release drugs locally to inflamed sites of intestine, contributing to superior therapeutic outcomes. Such strategy could also inspire the development of numerous hierarchical structured vehicles by other porous nanoparticles and stimuli-responsive materials for the local delivery of various drugs to treat plenty of inflammatory gastrointestinal diseases, including IBD, gastrointestinal cancers and viral infections.


Assuntos
Anti-Inflamatórios/administração & dosagem , Budesonida/administração & dosagem , Preparações de Ação Retardada/química , Doenças Inflamatórias Intestinais/tratamento farmacológico , Intestinos/efeitos dos fármacos , Silício/química , Administração Oral , Animais , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/uso terapêutico , Budesonida/farmacocinética , Budesonida/uso terapêutico , Linhagem Celular , Sistemas de Liberação de Medicamentos , Humanos , Ácido Hialurônico/análogos & derivados , Concentração de Íons de Hidrogênio , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Intestinos/imunologia , Intestinos/patologia , Masculino , Camundongos Endogâmicos C57BL , Nanopartículas/química , Polímeros/química , Porosidade
15.
Nano Lett ; 18(2): 1448-1453, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29382198

RESUMO

Porous silicon nanoparticles (PSiNPs) and gold nanorods (AuNRs) can be used as biocompatible nanocarriers for delivery of therapeutics but undesired leakage makes them inefficient. By encapsulating the PSiNPs and AuNRs in a hydrogel shell, we create a biocompatible functional nanocarrier that enables sustained release of therapeutics. Here, we report the fabrication of AuNRs-conjugated PSi nanoparticles (AuNRsPSiNPs) through two-step chemical reaction for high-capacity loading of hydrophobic and hydrophilic therapeutics with photothermal property. Furthermore, using water-in-oil microemulsion templates, we encapsulate the AuNRsPSiNPs within a calcium alginate hydrogel nanoshell, creating a versatile biocompatible nanocarrier to codeliver therapeutics for biomedical applications. We find that the functionalized nanohydrogel effectively controls the release rate of the therapeutics while maintaining a high loading efficiency and tunable loading ratios. Notably, combinations of therapeutics coloaded in the functionalized nanohydrogels significantly enhance inhibition of multidrug resistance through synergism and promote faster cancer cell death when combined with photothermal therapy. Moreover, the AuNRs can mediate the conversion of near-infrared laser radiation into heat, increasing the release of therapeutics as well as thermally inducing cell damage to promote faster cancer cell death. Our AuNRsPSiNPs functionalized calcium alginate nanohydrogel holds great promise for photothermal combination therapy and other advanced biomedical applications.

16.
Int J Pharm ; 536(1): 241-250, 2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-29195917

RESUMO

Bone tissue engineering is considered an alternative approach for conventional strategies available to treat bone defects. In this study, we have developed bone scaffolds composed of hydroxyapaptite (HAp), gelatin and mesoporous silica, all recognized as promising materials in bone tissue engineering due to favorable biocompatibility, osteoconductivity and drug delivery potential, respectively. These materials were coupled with conductive polypyrrole (PPy) polymer to create a novel bone scaffold for regenerative medicine. Conductive and non-conductive scaffolds were made by slurry casting method and loaded with a model antibiotic, vancomycin (VCM). Their properties were compared in different experiments in which scaffolds containing PPy showed good mechanical properties, higher protein adsorption and higher percentage of VCM release over a long duration of time compared to non-conductive scaffolds. Osteoblast cells were perfectly immersed into the gelatin matrix and remained viable for 14 days. Overall, new conductive composite bone scaffolds were created and the obtained results strongly verified the applicability of this conductive scaffold in drug delivery, encouraging its further development in tissue engineering applications.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Polímeros/química , Pirróis/química , Dióxido de Silício/química , Vancomicina/química , Animais , Materiais Biocompatíveis/química , Osso e Ossos/efeitos dos fármacos , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Durapatita/química , Condutividade Elétrica , Gelatina/química , Camundongos , Osteoblastos/efeitos dos fármacos , Engenharia Tecidual/métodos , Alicerces Teciduais
17.
Int J Pharm ; 533(1): 156-168, 2017 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-28963013

RESUMO

Lipid-polymer hybrid nanoparticles (LPHNPs) are emerging platforms for drug delivery applications. In the present study, methotrexate loaded LPHNPs consisted of PLGA and Lipoid S100 were fabricated by employing a single-step modified nanoprecipitation method combined with self-assembly. A three factor, three level Box Behnken design using Design-Expert® software was employed to access the influence of three independent variables on the particle size, drug entrapment and percent drug release. The optimized formulation was selected through numeric optimization approach. The results were supported with the ANOVA analysis, regression equations and response surface plots. Transmission electron microscope images indicated the nanosized and spherical shape of the LPHNPs with fair size distribution. The nanoparticles ranged from 176 to 308nm, which increased with increased polymer concentration. The increase in polymer and lipid concentration also increased the drug entrapment efficiency. The in vitro drug release was in range 70.34-91.95% and the release mechanism follow the Higuchi model (R2=0.9888) and Fickian diffusion (n<0.5). The in vitro cytotoxicity assay and confocal microscopy of the optimized formulation demonstrate the good safety and better internalization of the LPHNPs. The cell antiproliferation showed the spatial and controlled action of the nanoformulation as compared to the plain drug solution. The results suggest that LPHNPs can be a promising delivery system envisioned to safe, stable and potentially controlled delivery of methotrexate to the cancer cells to achieve better therapeutic outcomes.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Metotrexato/administração & dosagem , Nanopartículas/administração & dosagem , Antimetabólitos Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Liberação Controlada de Fármacos , Humanos , Ácido Láctico/administração & dosagem , Ácido Láctico/química , Metotrexato/química , Nanopartículas/química , Tamanho da Partícula , Fosfolipídeos/administração & dosagem , Fosfolipídeos/química , Ácido Poliglicólico/administração & dosagem , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
18.
Bioconjug Chem ; 28(6): 1639-1648, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28557453

RESUMO

The encapsulation of drugs to nanoparticles may offer a solution for targeted delivery. Here, we set out to engineer a self-assembling targeting ligand by combining the functional properties of human transferrin and fungal hydrophobins in a single fusion protein. We showed that human transferrin can be expressed in Nicotiana benthamiana plants as a fusion with Trichoderma reesei hydrophobins HFBI, HFBII, or HFBIV. Transferrin-HFBIV was further expressed in tobacco BY-2 suspension cells. Both partners of the fusion protein retained their functionality; the hydrophobin moiety enabled migration to a surfactant phase in an aqueous two-phase system, and the transferrin moiety was able to reversibly bind iron. Coating porous silicon nanoparticles with the fusion protein resulted in uptake of the nanoparticles in human cancer cells. This study provides a proof-of-concept for the functionalization of hydrophobin coatings with transferrin as a targeting ligand.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Proteínas Recombinantes de Fusão/metabolismo , Linhagem Celular Tumoral , Proteínas Fúngicas/genética , Humanos , Nanopartículas/uso terapêutico , Neoplasias/terapia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Nicotiana/metabolismo , Transferrina/genética
19.
Nanomedicine (Lond) ; 12(12): 1401-1420, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28524813

RESUMO

AIM: To evaluate the chemotherapeutic potential of a novel multifunctional nanocomposite encapsulating both porous silicon (PSi) and gold (Au) nanoparticles in a polymeric nanocomplex. MATERIALS & METHODS: The nanocomposite was physicochemically characterized and evaluated in vitro for biocompatibility, cellular internalization, endosomolytic properties, cytoplasmatic drug delivery and chemotherapeutic efficacy. RESULTS: The nanocomposites were successfully produced and exhibited adequate physicochemical properties and superior in vitro cyto- and hemocompatibilities. The encapsulation of PSi nanoparticles in the nanocomplexes significantly enhanced their cellular internalization and enabled their endosomal escape, resulting in the efficient cytoplasmic delivery of these nanosystems. Sorafenib-loaded nanocomposites showed a potent in vitro antiproliferative effect on MDA-MB-231 breast cancer cells. CONCLUSION: The multifunctional nanocomposite herein presented exhibits great potential as a chemotherapeutic nanoplatform.

20.
ACS Appl Mater Interfaces ; 9(16): 13976-13987, 2017 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-28383881

RESUMO

Hypoxia occurs in most solid tumors, and it has been shown to be an independent prognostic indicator of a poor clinical outcome for patients with various cancers. Therefore, constructing a nanosystem specifically targeting cancer cells under hypoxia conditions is a promising approach for cancer therapy. Herein, we develop a porous silicon (PSi)-based nanosystem for targeted cancer therapy. VD11-4-2, a novel inhibitor for carbonic anhydrase IX (CA IX), is anchored on PSi particles (VD-PSi). As CA IX is mainly expressed on the cancer cell membrane under hypoxia condition, this nanocomplex inherits a strong affinity toward hypoxic human breast adenocarcinoma (MCF-7) cells; thus, a better killing efficiency for the hypoxia-induced drug resistance cancer cell is observed. Furthermore, the release of doxorubicin (DOX) from VD-PSi showed pH dependence, which is possibly due to the hydrogen-bonding interaction between DOX and VD11-4-2. The fluorescence resonance energy transfer effect between DOX and VD11-4-2 is observed and applied for monitoring the DOX release intracellularly. Protein inhibition and binding assays showed that VD-PSi binds and inhibits CA IX. Overall, we developed a novel nanosystem inheriting several advantageous properties, which has great potential for targeted treatment of cancer cells under hypoxic conditions.


Assuntos
Anidrase Carbônica IX/química , Antígenos de Neoplasias , Hipóxia Celular , Linhagem Celular Tumoral , Doxorrubicina , Humanos , Silício
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA