Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.178
Filtrar
Más filtros











Intervalo de año de publicación
1.
ACS Nano ; 18(28): 18211-18229, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38946122

RESUMEN

Transarterial chemoembolization (TACE), the mainstay treatment of unresectable primary liver cancer that primarily employs nondegradable drug-loaded embolic agents to achieve synergistic vascular embolization and locoregional chemotherapy effects, suffers from an inferior drug burst behavior lacking long-term drug release controllability that severely limits the TACE efficacy. Here we developed gelatin-based drug-eluting microembolics grafted with nanosized poly(acrylic acid) serving as a biodegradable ion-exchange platform that leverages a counterion condensation effect to achieve high-efficiency electrostatic drug loading with electropositive drugs such as doxorubicin (i.e., drug loading capacity >34 mg/mL, encapsulation efficiency >98%, and loading time <10 min) and an enzymatic surface-erosion degradation pattern (∼2 months) to offer sustained locoregional pharmacokinetics with long-lasting deep-tumor retention capability for TACE treatment. The microembolics demonstrated facile microcatheter deliverability in a healthy porcine liver embolization model, superior tumor-killing capacity in a rabbit VX2 liver cancer embolization model, and stabilized extravascular drug penetration depth (>3 mm for 3 months) in a rabbit ear embolization model. Importantly, the microembolics finally exhibited vessel remodeling-induced permanent embolization with minimal inflammation responses after complete degradation. Such a biodegradable ion-exchange drug carrier provides an effective and versatile strategy for enhancing long-term therapeutic responses of various local chemotherapy treatments.


Asunto(s)
Quimioembolización Terapéutica , Doxorrubicina , Animales , Quimioembolización Terapéutica/métodos , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Conejos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/tratamiento farmacológico , Porcinos , Resinas Acrílicas/química , Polielectrolitos/química , Portadores de Fármacos/química , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/farmacocinética , Gelatina/química , Nanopartículas/química , Humanos , Liberación de Fármacos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación
2.
Int J Nanomedicine ; 19: 6829-6843, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39005958

RESUMEN

Background: With the rapid development of nanotechnology, constructing a multifunctional nanoplatform that can deliver various therapeutic agents in different departments and respond to endogenous/exogenous stimuli for multimodal synergistic cancer therapy remains a major challenge to address the inherent limitations of chemotherapy. Methods: Herein, we synthesized hollow mesoporous Prussian Blue@zinc phosphate nanoparticles to load glucose oxidase (GOx) and DOX (designed as HMPB-GOx@ZnP-DOX NPs) in the non-identical pore structures of their HMPB core and ZnP shell, respectively, for photothermally augmented chemo-starvation therapy. Results: The ZnP shell coated on the HMPB core, in addition to providing space to load DOX for chemotherapy, could also serve as a gatekeeper to protect GOx from premature leakage and inactivation before reaching the tumor site because of its degradation characteristics under mild acidic conditions. Moreover, the loaded GOx can initiate starvation therapy by catalyzing glucose oxidation while causing an upgradation of acidity and H2O2 levels, which can also be used as forceful endogenous stimuli to trigger smart delivery systems for therapeutic applications. The decrease in pH can improve the pH-sensitivity of drug release, and O2 can be supplied by decomposing H2O2 through the catalase-like activity of HMPBs, which is beneficial for relieving the adverse conditions of anti-tumor activity. In addition, the inner HMPB also acts as a photothermal agent for photothermal therapy and the generated hyperthermia upon laser irradiation can serve as an external stimulus to further promote drug release and enzymatic activities of GOx, thereby enabling a synergetic photothermally enhanced chemo-starvation therapy effect. Importantly, these results indicate that HMPB-GOx@ZnP-DOX NPs can effectively inhibit tumor growth by 80.31% and exhibit no obvious systemic toxicity in mice. Conclusion: HMPB-GOx@ZnP-DOX NPs can be employed as potential theranostic agents that incorporate multiple therapeutic modes to efficiently inhibit tumors.


Asunto(s)
Doxorrubicina , Ferrocianuros , Glucosa Oxidasa , Fosfatos , Terapia Fototérmica , Compuestos de Zinc , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Animales , Glucosa Oxidasa/química , Glucosa Oxidasa/farmacología , Ratones , Ferrocianuros/química , Ferrocianuros/farmacología , Humanos , Compuestos de Zinc/química , Fosfatos/química , Fosfatos/farmacología , Terapia Fototérmica/métodos , Porosidad , Nanopartículas/química , Línea Celular Tumoral , Liberación de Fármacos , Ratones Endogámicos BALB C , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Portadores de Fármacos/química
3.
ACS Appl Bio Mater ; 7(7): 4542-4552, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38957152

RESUMEN

Surface-engineered gold nanoparticles have been considered as versatile systems for theranostics applications. Moreover, surface covering or stabilizing agents on gold nanoparticles especially gold nanobipyramids (AuNBPs) provides an extra space for cargo molecules entrapment. However, it is not well studied yet and also the preparation of AuNBPs still remains dependent largely on cetyltrimethylammonium bromide (CTAB), a cytotoxic surfactant. Therefore, the direct use of CTAB stabilized nanoparticles is not recommended for cancer theranostics applications. Herein, we address an approach of dodecyl ethyl dimethylammonium bromide (DMAB) as biocompatible structure directing agent for AuNBPs, which also accommodate anticancer drug doxorubicin (45%), an additional chemotherapeutics agent. Upon near-infrared light (NIR, 808 nm) exposure, engineered AuNBPs exhibit (i) better phototransduction (51 °C) due to NIR absorption ability (650-900 nm), (ii) photo triggered drug release (more than 80%), and (iii) synergistic chemophototherapy for breast cancer cells. Drug release response has been evaluated in tumor microenvironment conditions (84% in acidic pH and 80% at high GSH) due to protonation and high affinity of thiol binding with AuNBPs followed by DMAB replacement. Intracellular glutathione (GSH, 5-7.5 mM) replaces DMAB from AuNBPs, which cause easy aggregation of nanoparticles as corroborated by colorimetric shifts, suggesting their utilization as a molecular sensing probe of early stage cancer biomarkers. Our optimized recipe yield is monodisperse DMAB-AuNBPs with ∼90% purity even at large scales (500 mL volume per batch). DMAB-AuNBPs show better cell viability (more than 90%) across all concentrations (5-500 ug/mL) when directly compared to CTAB-AuNBPs (less than 10%). Our findings show the potential of DMAB-AuNBPs for early stage cancer detection and theranostics applications.


Asunto(s)
Doxorrubicina , Ensayos de Selección de Medicamentos Antitumorales , Oro , Nanopartículas del Metal , Tamaño de la Partícula , Oro/química , Oro/farmacología , Humanos , Doxorrubicina/farmacología , Doxorrubicina/química , Nanopartículas del Metal/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Ensayo de Materiales , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Liberación de Fármacos , Estructura Molecular
4.
Molecules ; 29(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38999141

RESUMEN

Gemcitabine is a widely used antimetabolite drug of pyrimidine structure, which can exist as a free-base molecular form (Gem). The encapsulated forms of medicinal drugs are of interest for delayed and local drug release. We utilized, for the first time, a novel approach of mechano-chemistry by liquid-assisted grinding (LAG) to encapsulate Gem on a "matrix" of porphyrin aluminum metal-organic framework Al-MOF-TCPPH2 (compound 2). The chemical bonding of Gem to compound 2 was studied by ATR-FTIR spectroscopy and powder XRD. The interaction involves the C=O group of Gem molecules, which indicates the formation of the encapsulation complex in the obtained composite. Further, the delayed release of Gem from the composite was studied to phosphate buffered saline (PBS) at 37 °C using an automated drug dissolution apparatus equipped with an autosampler. The concentration of the released drug was determined by HPLC-UV analysis. The composite shows delayed release of Gem due to the bonded form and constant concentration thereafter, while pure Gem shows quick dissolution in less than 45 min. Delayed release of Gem drug from the composite follows the kinetic pseudo-first-order rate law. Further, for the first time, the mechanism of delayed release of Gem was assessed by the variable stirring speed of drug release media, and kinetic rate constant k was found to decrease when stirring speed is decreased (diffusion control). Finally, the prolonged time scale of toxicity of Gem to pancreatic cancer PANC-1 cells was studied by continuous measurements of proliferation (growth) for 6 days, using the xCELLigence real-time cell analyzer (RTCA), for the composite vs. pure drug, and their differences indicate delayed drug release. Aluminum metal-organic frameworks are new and promising materials for the encapsulation of gemcitabine and related small-molecule antimetabolites for controlled delayed drug release and potential use in drug-eluting implants.


Asunto(s)
Aluminio , Preparaciones de Acción Retardada , Desoxicitidina , Liberación de Fármacos , Gemcitabina , Estructuras Metalorgánicas , Neoplasias Pancreáticas , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/farmacología , Estructuras Metalorgánicas/química , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Línea Celular Tumoral , Aluminio/química , Preparaciones de Acción Retardada/farmacología , Preparaciones de Acción Retardada/química , Porfirinas/química , Porfirinas/farmacología , Supervivencia Celular/efectos de los fármacos , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/química
5.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000306

RESUMEN

The unique structure of G4.0 PAMAM dendrimers allows a drug to be enclosed in internal spaces or immobilized on the surface. In the conducted research, the conditions for the formation of the active G4.0 PAMAM complex with doxorubicin hydrochloride (DOX) were optimized. The physicochemical properties of the system were monitored using dynamic light scattering (DLS), circular dichroism (CD), and fluorescence spectroscopy. The Quartz Crystal Microbalance with Dissipation Monitoring (QCM-D) method was chosen to determine the preferential conditions for the complex formation. The highest binding efficiency of the drug to the cationic dendrimer was observed under basic conditions when the DOX molecule was deprotonated. The decrease in the zeta potential of the complex confirms that DOX immobilizes through electrostatic interaction with the carrier's surface amine groups. The binding constants were determined from the fluorescence quenching of the DOX molecule in the presence of G4.0 PAMAM. The two-fold way of binding doxorubicin in the structure of dendrimers was visible in the Isothermal calorimetry (ITC) isotherm. Fluorescence spectra and release curves identified the reversible binding of DOX to the nanocarrier. Among the selected cancer cells, the most promising anticancer activity of the G4.0-DOX complex was observed in A375 malignant melanoma cells. Moreover, the preferred intracellular location of the complexes concerning the free drug was found, which is essential from a therapeutic point of view.


Asunto(s)
Dendrímeros , Doxorrubicina , Dendrímeros/química , Doxorrubicina/química , Doxorrubicina/farmacología , Humanos , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Supervivencia Celular/efectos de los fármacos
6.
Sci Adv ; 10(28): eadn0960, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996025

RESUMEN

Celastrol (CEL), an active compound isolated from the root of Tripterygium wilfordii, exhibits broad anticancer activities. However, its poor stability, narrow therapeutic window and numerous adverse effects limit its applications in vivo. In this study, an adenosine triphosphate (ATP) activatable CEL-Fe(III) chelate was designed, synthesized, and then encapsulated with a reactive oxygen species (ROS)-responsive polymer to obtain CEL-Fe nanoparticles (CEL-Fe NPs). In normal tissues, CEL-Fe NPs maintain structural stability and exhibit reduced systemic toxicity, while at the tumor site, an ATP-ROS-rich tumor microenvironment, drug release is triggered by ROS, and antitumor potency is restored by competitive binding of ATP. This intelligent CEL delivery system improves the biosafety and bioavailability of CEL for cancer therapy. Such a CEL-metal chelate strategy not only mitigates the challenges associated with CEL but also opens avenues for the generation of CEL derivatives, thereby expanding the therapeutic potential of CEL in clinical settings.


Asunto(s)
Adenosina Trifosfato , Triterpenos Pentacíclicos , Profármacos , Especies Reactivas de Oxígeno , Triterpenos Pentacíclicos/farmacología , Triterpenos Pentacíclicos/química , Profármacos/química , Profármacos/farmacología , Adenosina Trifosfato/metabolismo , Humanos , Animales , Especies Reactivas de Oxígeno/metabolismo , Ratones , Línea Celular Tumoral , Triterpenos/química , Triterpenos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Quelantes/química , Quelantes/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Liberación de Fármacos , Nanopartículas/química , Ensayos Antitumor por Modelo de Xenoinjerto , Compuestos Férricos/química
7.
Eur J Pharm Biopharm ; 201: 114386, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38950717

RESUMEN

The goal of the study was to fabricate folic acid functionalized docetaxel (DOC)/erlotinib (ERL)-loaded solid lipid nanoparticles (SLNs) to synergistically increase the anticancer activity against triple-negative breast cancer. DOC/ERL-SLNs were prepared by the high shear homogenization - ultrasound dispersion method (0.1 % w/v for DOC, and 0.3 %w/v for ERL) and optimized using Plackett Burman Design (PBD) followed by Box Behnken Design (BBD). The optimized SLNs demonstrated particle size < 200 nm, PDI < 0.35, and negative zeta potential with entrapment and loading efficiency of ∼80 and ∼4 %, respectively. The SLNs and folic acid functionalized SLNs (FA-SLNs) showed sustained release for both drugs, followed by Higuchi and Korsemeyer-Peppas drug release models, respectively. Further, the in vitro pH-stat lipolysis model demonstrated an approximately 3-fold increase in the bioaccessibility of drugs from SLNs compared to suspension. The TEM images revealed the spherical morphology of the SLNs. DOC/ERL loaded SLNs showed dose- and time-dependent cytotoxicity and exhibited a synergism at a molar ratio of 1:3 in TNBC with a combination index of 0.35 and 0.37, respectively. FA-DOC/ERL-SLNs showed enhanced anticancer activity as evidenced by MMP and ROS assay and further inhibited the colony-forming ability and the migration capacity of TNBC cells. Conclusively, the study has shown that SLNs are encouraging systems to improve the pharmaceutical attributes of poorly bioavailable drugs.


Asunto(s)
Docetaxel , Liberación de Fármacos , Sinergismo Farmacológico , Clorhidrato de Erlotinib , Lípidos , Nanopartículas , Tamaño de la Partícula , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Docetaxel/administración & dosificación , Docetaxel/farmacología , Docetaxel/farmacocinética , Humanos , Nanopartículas/química , Clorhidrato de Erlotinib/administración & dosificación , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/farmacocinética , Línea Celular Tumoral , Femenino , Lípidos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Portadores de Fármacos/química , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/química , Liposomas
8.
J Mol Model ; 30(8): 261, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38985223

RESUMEN

CONTEXT: Multiwalled carbon nanotubes (MWCNTs) functionalized with lysine via 1,3-dipolar cycloaddition and conjugated to galactose or mannose are potential nanocarriers that can effectively bind to the lectin receptor in MDA-MB-231 or MCF-7 breast cancer cells. In this work, a method based on molecular dynamics (MD) simulation was used to predict the interaction of these functionalized MWCNTs with doxorubicin and obtain structural evidence that allows a better understanding of the drug loading and release process. The MD simulations showed that while doxorubicin only interacted with pristine MWCNTs through π-π stacking interactions, functionalized MWCNTs were also able to establish hydrogen bonds, suggesting that the functionalized groups improve doxorubicin loading. Moreover, the elevated adsorption levels observed for functionalized nanotubes further support this enhancement in loading efficiency. MD simulations also shed light on the intratumoral pH-specific release of doxorubicin from functionalized MWCNTs, which is induced by protonation of the daunosamine moiety. The simulations show that this change in protonation leads to a lower absorption of doxorubicin to the MWCNTs. The MD studies were then experimentally validated, where functionalized MWCNTs showed improved dispersion in aqueous medium compared to pristine MWCNTs and, in agreement with the computational predictions, increased drug loading capacity. Doxorubicin-loaded functionalized MWCNTs demonstrated specific release of doxorubicin in tumor microenvironment (pH = 5.0) with negligible release in the physiological pH (pH = 7.4). Furthermore, doxorubicin-free MWNCT nanoformulations exhibited insignificant cytotoxicity. The experimental studies yielded nearly identical results to the MD studies, underlining the usefulness of the method. Our functionalized MWCNTs represent promising non-toxic nanoplatforms with enhanced aqueous dispersibility and the potential for conjugation with ligands for targeted delivery of anti-cancer drugs to breast cancer cells. METHODS: The computational model of a pristine carbon nanotube was created with the buildCstruct 1.2 Python script. The lysinated functionalized groups were added with PyMOL and VMD. The carbon nanotubes and doxorubicin molecules were parameterized using the general AMBER force field, and RESP charges were determined using Gaussian 09. Molecular dynamics simulations were carried out with the AMBER 20 software package. Adsorption levels were calculated using the water-shell function of cpptraj. Cytotoxicity was evaluated via a MTT assay using MDA-MB-231 and MCF-7 breast cancer cells. Drug uptake of doxorubicin and doxorubicin-loaded MWCNTs was measured by fluorescence microscopy.


Asunto(s)
Doxorrubicina , Simulación de Dinámica Molecular , Nanotubos de Carbono , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Nanotubos de Carbono/química , Humanos , Lisina/química , Portadores de Fármacos/química , Células MCF-7 , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Línea Celular Tumoral , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/administración & dosificación
9.
AAPS PharmSciTech ; 25(6): 153, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961013

RESUMEN

Despite ongoing advances in cancer therapy, the results for the treatment of breast cancer are not satisfactory. The advent of nanotechnology promises to be an essential tool to improve drug delivery effectiveness in cancer therapy. Nanotechnology provides an opportunity to enhance the treatment modality by preventing degradation, improving tumour targeting, and controlling drug release. Recent advances have revealed several strategies to prevent cancer metastasis using nano-drug delivery systems (NDDS). These strategies include the design of appropriate nanocarriers loaded with anti-cancer drugs that target the optimization of physicochemical properties, modulate the tumour microenvironment, and target biomimetic techniques. Nanocarriers have emerged as a preferential approach in the chemotropic treatment for breast cancer due to their pivotal role in safeguarding the therapeutic agents against degradation. They facilitate efficient drug concentration in targeted cells, surmount the resistance of drugs, and possess a small size. Nevertheless, these nanocarrier(s) have some limitations, such as less permeability across the barrier and low bioavailability of loaded drugs. To overcome these challenges, integrating external stimuli has been employed, encompassing infrared light, thermal stimulation, microwaves, and X-rays. Among these stimuli, ultrasound-triggered nanocarriers have gained significant attention due to their cost-effectiveness, non-invasive nature, specificity, ability to penetrate tissues, and capacity to deliver elevated drug concentrations to intended targets. This article comprehensively reviews recent advancements in different nanocarriers for breast cancer chemotherapy. It also delves into the associated hurdles and offers valuable insights into the prospective directions for this innovative field.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Portadores de Fármacos , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Humanos , Portadores de Fármacos/química , Antineoplásicos/administración & dosificación , Femenino , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos , Animales , Liberación de Fármacos , Nanotecnología/métodos
10.
Mikrochim Acta ; 191(8): 447, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38963544

RESUMEN

An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).


Asunto(s)
Cobre , Doxorrubicina , Glucosa Oxidasa , Ácido Hialurónico , Estructuras Metalorgánicas , Microambiente Tumoral , Zeolitas , Cobre/química , Doxorrubicina/farmacología , Doxorrubicina/química , Microambiente Tumoral/efectos de los fármacos , Glucosa Oxidasa/química , Glucosa Oxidasa/metabolismo , Humanos , Zeolitas/química , Animales , Estructuras Metalorgánicas/química , Ácido Hialurónico/química , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Línea Celular Tumoral , Ratones , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/química , Neoplasias/tratamiento farmacológico , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Imidazoles
11.
Drug Deliv ; 31(1): 2372277, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38952058

RESUMEN

Skin melanoma is considered the most dangerous form of skin cancer due to its association with high risk of metastasis, high mortality rate and high resistance to different treatment options. Genistein is a natural isoflavonoid with known chemotherapeutic activity. Unfortunately, it has low bioavailability due to its poor aqueous solubility and excessive metabolism. In the current study, genistein was incorporated into transferosomal hydrogel to improve its bioavailability. The prepared transferosomal formulations were characterized regarding: particle size; polydispersity index; zeta potential; encapsulation efficiency; TEM; FTIR; DSC; XRD; in vitro drug release; viscosity; pH; ex vivo anti-tumor activity on 3D skin melanoma spheroids and 1-year stability study at different storage temperatures. The optimized formulation has high encapsulation efficiency with an excellent particle size that will facilitate its penetration through the skin. The transfersomes have a spherical shape with sustained drug release profile. The anti-tumor activity evaluation of genistein transfersome revealed that genistein is a potent chemotherapeutic agent with enhanced penetration ability through the melanoma spheroids when incorporated into transfersomes. Stability study results demonstrate the high physical and chemical stability of our formulations. All these outcomes provide evidence that our genistein transferosomal hydrogel is a promising treatment option for skin melanoma.


Asunto(s)
Liberación de Fármacos , Genisteína , Hidrogeles , Melanoma , Tamaño de la Partícula , Neoplasias Cutáneas , Genisteína/administración & dosificación , Genisteína/farmacología , Genisteína/farmacocinética , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Humanos , Hidrogeles/química , Sistemas de Liberación de Medicamentos/métodos , Línea Celular Tumoral , Estabilidad de Medicamentos , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Solubilidad , Portadores de Fármacos/química , Química Farmacéutica , Viscosidad , Disponibilidad Biológica , Administración Cutánea , Esferoides Celulares/efectos de los fármacos
12.
Drug Deliv ; 31(1): 2372285, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38952133

RESUMEN

In this study, chitosan low molecular weight (LCH) and chitosan medium molecular weight (MCH) were employed to encapsulate a yarrow extract rich in chlorogenic acid and dicaffeoylquinic acids (DCQAs) that showed antiproliferative activity against colon adenocarcinoma cells. The design of CH micro/nanoparticles to increase the extract colon delivery was carried out by using two different techniques: ionic gelation and spray drying. Ionic gelation nanoparticles obtained were smaller and presented higher yields values than spray-drying microparticles, but spray-drying microparticles showed the best performance in terms of encapsulation efficiency (EE) (> 94%), also allowing the inclusion of a higher quantity of extract. Spray-drying microparticles designed using LCH with an LCH:extract ratio of 6:1 (1.25 mg/mL) showed a mean diameter of 1.31 ± 0.21 µm and EE values > 93%, for all phenolic compounds studied. The release profile of phenolic compounds included in this formulation, at gastrointestinal pHs (2 and 7.4), showed for most of them a small initial release, followed by an increase at 1 h, with a constant release up to 3 h. Chlorogenic acid presented the higher release values at 3 h (56.91% at pH 2; 44.45% at pH 7.4). DCQAs release at 3 h ranged between 9.01- 40.73%, being higher for 1,5- and 3,4-DCQAs. After gastrointestinal digestion, 67.65% of chlorogenic and most DCQAs remained encapsulated. Therefore, spray-drying microparticles can be proposed as a promising vehicle to increase the colon delivery of yarrow phenolics compounds (mainly chlorogenic acid and DCQAs) previously described as potential agents against colorectal cancer.


Asunto(s)
Achillea , Proliferación Celular , Quitosano , Ácido Clorogénico , Neoplasias Colorrectales , Nanopartículas , Tamaño de la Partícula , Extractos Vegetales , Quitosano/química , Humanos , Extractos Vegetales/farmacología , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Achillea/química , Ácido Clorogénico/farmacología , Ácido Clorogénico/administración & dosificación , Ácido Clorogénico/química , Nanopartículas/química , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Línea Celular Tumoral , Ácido Quínico/análogos & derivados , Ácido Quínico/farmacología , Ácido Quínico/química , Ácido Quínico/administración & dosificación , Liberación de Fármacos , Sistemas de Liberación de Medicamentos/métodos , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Colon/efectos de los fármacos , Colon/metabolismo , Portadores de Fármacos/química , Peso Molecular
13.
Int J Nanomedicine ; 19: 6519-6546, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957181

RESUMEN

Background: Salidroside (SAL) is the most effective component of Rhodiola rosea, a traditional Chinese medicine. Cryptotanshinone (CT) is the main fat-soluble extract of Salvia miltiorrhiza, exhibiting considerable potential for application in osteogenesis. Herein, a polycaprolactone/gelatin nanofiber membrane loaded with CT and SAL (PSGC membrane) was successfully fabricated via coaxial electrospinning and characterized. Methods and Results: This membrane capable of sustained and controlled drug release was employed in this study. Co-culturing the membrane with bone marrow mesenchymal stem cells and human umbilical vein endothelial cells revealed excellent biocompatibility and demonstrated osteogenic and angiogenic capabilities. Furthermore, drug release from the PSGC membrane activated the Wnt/ß-catenin signaling pathway and promoted osteogenic differentiation and vascularization. Evaluation of the membrane's vascularization and osteogenic capacities involved transplantation onto a rat's subcutaneous area and assessing rat cranium defects for bone regeneration, respectively. Microcomputed tomography, histological tests, immunohistochemistry, and immunofluorescence staining confirmed the membrane's outstanding angiogenic capacity two weeks post-operation, with a higher incidence of osteogenesis observed in rat cranial defects eight weeks post-surgery. Conclusion: Overall, the SAL- and CT-loaded coaxial electrospun nanofiber membrane synergistically enhances bone repair and regeneration.


Asunto(s)
Gelatina , Glucósidos , Células Endoteliales de la Vena Umbilical Humana , Células Madre Mesenquimatosas , Nanofibras , Neovascularización Fisiológica , Osteogénesis , Fenantrenos , Fenoles , Poliésteres , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Animales , Nanofibras/química , Gelatina/química , Poliésteres/química , Glucósidos/química , Glucósidos/farmacología , Fenoles/química , Fenoles/farmacología , Fenantrenos/química , Fenantrenos/farmacología , Fenantrenos/farmacocinética , Fenantrenos/administración & dosificación , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Ratas , Masculino , Regeneración Ósea/efectos de los fármacos , Membranas Artificiales , Técnicas de Cocultivo , Liberación de Fármacos , Diferenciación Celular/efectos de los fármacos
14.
ACS Appl Mater Interfaces ; 16(27): 34669-34683, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38946103

RESUMEN

In this research, a novel MgSiO3 fiber membrane (MSFM) loaded with indocyanine green (ICG) and doxorubicin (DOX) was prepared. Because of MgSiO3's unique lamellar structure composed of a silicon-oxygen tetrahedron, magnesium ion (Mg2+) moves easily and can be further replaced with other cations. Therefore, because of the positively charged functional group of ICG, MSFM has a rather high drug loading for ICG. In addition, there is electrostatic attraction between DOX (a cationic drug) and ICG (an anionic drug). Hence, after loading ICG, more DOX can be adsorbed into MSFM because of electrostatic interaction. The ICG endows the MSFM outstanding photothermal therapy (PTT) performance, and DOX as a chemotherapeutic drug can restrain tumor growth. On the one hand, H+ exchanged with the positively charged DOX based on the MgSiO3 special lamellar structure. On the other hand, the thermal effect could break the electrostatic interaction between ICG and DOX. Based on the above two points, both tumor acidic microenvironment and photothermal effect can trigger DOX release. What's more, in vitro and in vivo antiosteosarcoma therapy evaluations displayed a superior synergetic PTT-chemotherapy anticancer treatment and excellent biocompatibility of DOX&ICG-MSFM. Finally, the MSFM was proven to greatly promote cell proliferation, differentiation, and bone regeneration performance in vitro and in vivo. Therefore, MSFM provides a creative perspective in the design of multifunctional scaffolds and shows promising applications in controlled drug delivery, antitumor performance, and osteogenesis.


Asunto(s)
Regeneración Ósea , Doxorrubicina , Verde de Indocianina , Osteosarcoma , Doxorrubicina/química , Doxorrubicina/farmacología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Regeneración Ósea/efectos de los fármacos , Animales , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacología , Ratones , Silicatos de Magnesio/química , Terapia Fototérmica , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Liberación de Fármacos
15.
ACS Appl Mater Interfaces ; 16(27): 34510-34523, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38946393

RESUMEN

Photoluminescence (PL) metal nanoclusters (NCs) have attracted extensive attention due to their excellent physicochemical properties, good biocompatibility, and broad application prospects. However, developing water-soluble PL metal NCs with a high quantum yield (QY) and high stability for visual drug delivery remains a great challenge. Herein, we have synthesized ultrabright l-Arg-ATT-Au/Ag NCs (Au/Ag NCs) with a PL QY as high as 73% and excellent photostability by heteroatom doping and surface rigidization in aqueous solution. The as-prepared Au/Ag NCs can maintain a high QY of over 61% in a wide pH range and various ionic environments as well as a respectable resistance to photobleaching. The results from structure characterization and steady-state and time-resolved spectroscopic analysis reveal that Ag doping into Au NCs not only effectively modifies the electronic structure and photostability but also significantly regulates the interfacial dynamics of the excited states and enhances the PL QY of Au/Ag NCs. Studies in vitro indicate Au/Ag NCs have a high loading capacity and pH-triggered release ability of doxorubicin (DOX) that can be visualized from the quenching and recovery of PL intensity and lifetime. Imaging-guided experiments in cancer cells show that DOX of Au/Ag NCs-DOX agents can be efficiently delivered and released in the nucleus with preferential accumulation in the nucleolus, facilitating deep insight into the drug action sites and pharmacological mechanisms. Moreover, the evaluation of anticancer activity in vivo reveals an outstanding suppression rate of 90.2% for mice tumors. These findings demonstrate Au/Ag NCs to be a superior platform for bioimaging and visual drug delivery in biomedical applications.


Asunto(s)
Doxorrubicina , Oro , Nanopartículas del Metal , Plata , Agua , Oro/química , Plata/química , Plata/farmacología , Humanos , Animales , Doxorrubicina/química , Doxorrubicina/farmacología , Nanopartículas del Metal/química , Ratones , Agua/química , Sistemas de Liberación de Medicamentos , Células HeLa , Portadores de Fármacos/química , Solubilidad , Liberación de Fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Luminiscencia
16.
Biomed Pharmacother ; 177: 117107, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38996708

RESUMEN

The development of new effective drugs to treat breast cancer remains a huge challenge. ABT-737 can inhibit Bcl-2 proteins to promote apoptosis. Resiquimod (R848) is a TLR7/8 agonist that is effective in modulating the immunosuppressive microenvironment. In this study, a codelivery system (TPGS/ABT+R848 NPs) based on D-α-tocopheryl poly (ethylene glycol) 1000 succinate as a potential drug delivery vector to codelivery ABT-737 and R848 was investigated. The size of TPGS/ABT+R848 NPs was 102.5 nm, the drug loading of ABT-737 and R848 was 30.6 % and 12.5 %, and the entrapment efficiency was 84.2 % and 23.7 %, respectively. The nanoparticles showed no significant change in particle size over 14 days. R848 and ABT-737 were released in co-loaded nanoparticles in sequential order. In vitro anti-tumor experiments, the IC50 value of TPGS/ABT+R848 NPs was 0.30 µg·mL-1, 34 times lower than that of free ABT-737. Animal experiments also verified that TPGS/ABT+R848 NPs could enhance the anti-tumor activity, and the tumor weight inhibition rate was 75.3 %. This study demonstrated that TPGS NPs loaded with ABT-737 and R848 have superior combination tumor therapeutic effects, and the co-loaded preparation is conducive to anti-tumor efficacy. The TPGS/ABT+R848 NPs could be a promising platform against breast cancer.


Asunto(s)
Compuestos de Bifenilo , Neoplasias de la Mama , Imidazoles , Nanopartículas , Nitrofenoles , Piperazinas , Sulfonamidas , Vitamina E , Sulfonamidas/farmacología , Sulfonamidas/administración & dosificación , Femenino , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Nitrofenoles/farmacología , Nitrofenoles/administración & dosificación , Humanos , Imidazoles/farmacología , Imidazoles/administración & dosificación , Piperazinas/farmacología , Piperazinas/administración & dosificación , Nanopartículas/química , Vitamina E/farmacología , Compuestos de Bifenilo/farmacología , Ratones , Ratones Endogámicos BALB C , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Tamaño de la Partícula , Liberación de Fármacos , Portadores de Fármacos/química , Ensayos Antitumor por Modelo de Xenoinjerto , Células MCF-7
17.
Int J Mol Sci ; 25(14)2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39063114

RESUMEN

The aim of this study was to examine the use of zwitterionic microparticles as new and efficient macromolecular supports for the sorption of an antibiotic (doxycycline hydrochloride, DCH) from aqueous solution. The effect of relevant process parameters of sorption, like dosage of microparticles, pH value, contact time, the initial concentration of drug and temperature, was evaluated to obtain the optimal experimental conditions. The sorption kinetics were investigated using Lagergren, Ho, Elovich and Weber-Morris models, respectively. The sorption efficiency was characterized by applying the Langmuir, Freundlich and Dubinin-Radushkevich isotherm models. The calculated thermodynamic parameters (ΔH, ΔS and ΔG) show that the sorption of doxycycline hydrochloride onto zwitterionic microparticles is endothermic, spontaneous and favorable at higher temperatures. The maximum identified sorption capacity value is 157.860 mg/g at 308 K. The Higuchi, Korsmeyer-Peppas, Baker-Lonsdale and Kopcha models are used to describe the release studies. In vitro release studies show that the release mechanism of doxycycline hydrochloride from zwitterionic microparticles is predominantly anomalous or non-Fickian diffusion. This study could provide the opportunity to expand the use of these new zwitterionic structures in medicine and water purification.


Asunto(s)
Betaína , Doxiciclina , Doxiciclina/química , Betaína/química , Cinética , Adsorción , Termodinámica , Concentración de Iones de Hidrógeno , Antibacterianos/química , Liberación de Fármacos , Temperatura , Microesferas
18.
J Control Release ; 372: 829-845, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38964471

RESUMEN

Cancer metastasis and recurrence are obstacles to successful treatment of aggressive cancer. To address this challenge, chemotherapy is indispensable as an essential part of comprehensive cancer treatment, particularly for subsequent therapy after surgical resection. However, small-molecule drugs for chemotherapy always cause inadequate efficacy and severe side effects against cancer metastasis and recurrence caused by lymph node metastases. Here, we developed doxorubicin-carried albumin nanocages (Dox-AlbCages) with appropriate particle sizes and pH/enzyme-responsive drug release for tumor and lymph node dual-targeted therapy by exploiting the inborn transport properties of serum albumin. Inspired by the protein-templated biomineralization and remote loading of doxorubicin into liposomes, we demonstrated the controlled synthesis of Dox-AlbCages via the aggregation or crystallization of doxorubicin and ammonium sulfate within albumin nanocages using a biomineralization strategy. Dox-AlbCages allowed efficient encapsulation of Dox in the core protected by the albumin corona shell, exhibiting favorable properties for enhanced tumor and lymph node accumulation and preferable cellular uptake for tumor-specific chemotherapy. Intriguingly, Dox-AlbCages effectively inhibited tumor growth and metastasis in orthotopic 4T1 breast tumors and prevented postsurgical tumor recurrence and lung metastasis. At the same time, Dox-AlbCages had fewer side effects than free Dox. This nanoplatform provides a facile strategy for designing tumor- and lymph node-targeted nanomedicines for suppressing cancer metastasis and recurrence.


Asunto(s)
Antibióticos Antineoplásicos , Doxorrubicina , Metástasis Linfática , Ratones Endogámicos BALB C , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Animales , Femenino , Línea Celular Tumoral , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapéutico , Liberación de Fármacos , Nanopartículas/administración & dosificación , Nanopartículas/química , Humanos , Ratones , Ganglios Linfáticos/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Sistemas de Liberación de Medicamentos , Albúminas/administración & dosificación , Albúminas/química , Portadores de Fármacos/química
19.
J Control Release ; 372: 810-828, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38968969

RESUMEN

Cancer is a serious threat to human health because of its high annual mortality rate. It has attracted significant attention in healthcare, and identifying effective strategies for the treatment and relief of cancer pain requires urgency. Drug delivery systems (DDSs) offer the advantages of excellent efficacy, low cost, and low toxicity for targeting drugs to tumor sites. In recent decades, copolymer carriers based on poly(phenylalanine) (PPhe) and poly(3,4-dihydroxy-L-phenylalanine) (PDopa) have been extensively investigated owing to their good biocompatibility, biodegradability, and controllable stimulus responsiveness, which have resulted in DDSs with loading and targeted delivery capabilities. In this review, we introduce the synthesis of PPhe and PDopa, highlighting the latest proposed synthetic routes and comparing the differences in drug delivery between PPhe and PDopa. Subsequently, we summarize the various applications of PPhe and PDopa in nanoscale-targeted DDSs, providing a comprehensive analysis of the drug release behavior based on different stimulus-responsive carriers using these two materials. In the end, we discuss the challenges and prospects of polypeptide-based DDSs in the field of cancer therapy, aiming to promote their further development to meet the growing demands for treatment.


Asunto(s)
Portadores de Fármacos , Humanos , Portadores de Fármacos/química , Animales , Sistemas de Liberación de Medicamentos , Péptidos/química , Péptidos/administración & dosificación , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Polímeros/química , Liberación de Fármacos , Fenilalanina/química , Fenilalanina/administración & dosificación , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Materiales Biocompatibles/química
20.
J Control Release ; 372: 874-884, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38977133

RESUMEN

Dexamethasone (DEX) has been demonstrated to inhibit the inflammatory corneal neovascularization (CNV). However, the therapeutic efficacy of DEX is limited by the poor bioavailability of conventional eye drops and the increased risk of hormonal glaucoma and cataract associated with prolonged and frequent usage. To address these limitations, we have developed a novel DEX-loaded, reactive oxygen species (ROS)-responsive, controlled-release nanogel, termed DEX@INHANGs. This advanced nanogel system is constructed by the formation of supramolecular host-guest complexes by cyclodextrin (CD) and adamantane (ADA) as a cross-linking force. The introduction of the ROS-responsive material, thioketal (TK), ensures the controlled release of DEX in response to oxidative stress, a characteristic of CNV. Furthermore, the nanogel's prolonged retention on the corneal surface for over 8 h is achieved through covalent binding of the integrin ß1 fusion protein, which enhances its bioavailability. Cytotoxicity assays demonstrated that DEX@INHANGs was not notably toxic to human corneal epithelial cells (HCECs). Furthermore, DEX@INHANGs has been demonstrated to effectively inhibit angiogenesis in vitro. In a rabbit model with chemically burned eyes, the once-daily topical application of DEX@INHANGs was observed to effectively suppress CNV. These results collectively indicate that the nanomedicine formulation of DEX@INHANGs may offer a promising treatment option for CNV, offering significant advantages such as reduced dosing frequency and enhanced patient compliance.


Asunto(s)
Neovascularización de la Córnea , Dexametasona , Especies Reactivas de Oxígeno , Animales , Conejos , Neovascularización de la Córnea/tratamiento farmacológico , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Humanos , Especies Reactivas de Oxígeno/metabolismo , Nanogeles/química , Preparaciones de Acción Retardada , Córnea/metabolismo , Córnea/efectos de los fármacos , Masculino , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/química , Línea Celular , Polietilenglicoles/química , Polietilenglicoles/administración & dosificación , Administración Oftálmica , Adamantano/administración & dosificación , Adamantano/análogos & derivados , Ciclodextrinas/química , Antiinflamatorios/administración & dosificación , Polietileneimina/química , Polietileneimina/administración & dosificación , Liberación de Fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA