Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 995
Filtrar
1.
Int J Nanomedicine ; 19: 8437-8461, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39170101

RESUMEN

Data published in 2020 by the International Agency for Research on Cancer (IARC) of the World Health Organization show that breast cancer (BC) has become the most common cancer globally, affecting more than 2 million women each year. The complex tumor microenvironment, drug resistance, metastasis, and poor prognosis constitute the primary challenges in the current diagnosis and treatment of BC. Magnetic iron oxide nanoparticles (MIONPs) have emerged as a promising nanoplatform for diagnostic tumor imaging as well as therapeutic drug-targeted delivery due to their unique physicochemical properties. The extensive surface engineering has given rise to multifunctionalized MIONPs. In this review, the latest advancements in surface modification strategies of MIONPs over the past five years are summarized and categorized as constrast agents and drug delivery platforms. Additionally, the remaining challenges and future prospects of MIONPs-based targeted delivery are discussed.


Asunto(s)
Neoplasias de la Mama , Nanopartículas Magnéticas de Óxido de Hierro , Humanos , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Nanopartículas Magnéticas de Óxido de Hierro/química , Sistemas de Liberación de Medicamentos/métodos , Propiedades de Superficie , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Animales , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Microambiente Tumoral/efectos de los fármacos
2.
Carbohydr Polym ; 343: 122455, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39174092

RESUMEN

Polysaccharides like hyaluronan (HA) and chondroitin sulfate (CS) are native of the brain's extracellular matrix crucial for myelination and brain maturation. Despite extensive research on HA and CS as drug delivery systems (DDS), their high water solubility limits their application as drug carriers. This study introduces an injectable DDS using aldehyde-modified hyaluronic acid (HAOX) hydrogel containing polyelectrolyte complexes (PEC) formed with calcium, gelatin, and either CS or aldehyde-modified CS (CSOX) to deliver minocycline for Multiple Sclerosis therapy. PECs with CSOX enable covalent crosslinking to HAOX, creating immobilized PECs (HAOX_PECOX), while those with CS remain unbound (HAOX_PECS). The in situ forming DDS can be administered via a 20 G needle, with rapid gelation preventing premature leakage. The system integrates into an implanted device for minocycline release through either Fickian or anomalous diffusion, depending on PEC immobilization. HAOX_PECOX reduced burst release by 88 %, with a duration of 127 h for 50 % release. The DDS exhibited an elastic modulus of 3800 Pa and a low swelling ratio (0-1 %), enabling precise control of minocycline release kinetics. Released minocycline reduced IL-6 secretion in the Whole Blood Monocytes Activation Test, suggesting that DDS formation may not alter the biological activity of the loaded drug.


Asunto(s)
Sulfatos de Condroitina , Portadores de Fármacos , Gelatina , Ácido Hialurónico , Hidrogeles , Minociclina , Polielectrolitos , Ácido Hialurónico/química , Gelatina/química , Sulfatos de Condroitina/química , Hidrogeles/química , Hidrogeles/farmacología , Minociclina/química , Minociclina/farmacología , Minociclina/administración & dosificación , Polielectrolitos/química , Humanos , Portadores de Fármacos/química , Liberación de Fármacos , Aldehídos/química , Animales , Sistemas de Liberación de Medicamentos/métodos , Interleucina-6/metabolismo
3.
Carbohydr Polym ; 343: 122486, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39174104

RESUMEN

Ophthalmic diseases can be presented as acute diseases like allergies, ocular infections, etc., or chronic ones that can be manifested as a result of systemic disorders, like diabetes mellitus, thyroid, rheumatic disorders, and others. Chitosan (CS) and its derivatives have been widely investigated as nanocarriers in the delivery of drugs, genes, and many biological products. The biocompatibility and biodegradability of CS made it a good candidate for ocular delivery of many ingredients, including immunomodulating agents, antibiotics, ocular hypertension medications, etc. CS-based nanosystems have been successfully reported to modulate ocular diseases by penetrating biological ocular barriers and targeting and controlling drug release. This review provides guidance to drug delivery formulators on the most recently published strategies that can enhance drug permeation to the ocular tissues in CS-based nanosystems, thus improving therapeutic effects through enhancing drug bioavailability. This review will highlight the main ocular barriers to drug delivery observed in the nano-delivery system. In addition, the CS physicochemical properties that contribute to formulation aspects are discussed. It also categorized the permeation enhancement strategies that can be optimized in CS-based nanosystems into four aspects: CS-related physicochemical properties, formulation components, fabrication conditions, and adopting a novel delivery system like implants, inserts, etc. as described in the published literature within the last ten years. Finally, challenges encountered in CS-based nanosystems and future perspectives are mentioned.


Asunto(s)
Quitosano , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Hidrogeles , Quitosano/química , Humanos , Hidrogeles/química , Animales , Sistemas de Liberación de Medicamentos/métodos , Portadores de Fármacos/química , Oftalmopatías/tratamiento farmacológico , Administración Oftálmica , Ojo/metabolismo , Ojo/efectos de los fármacos , Nanopartículas/química
4.
Carbohydr Polym ; 343: 122505, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39174110

RESUMEN

The development of thrombolytic drug carriers capable of thrombus-targeting, prolonged circulation time, intelligent responsive release, and the ability to inhibit thrombotic recurrences remains a promising but significant challenge. To tackle this, an artificial polysaccharide microvesicle drug delivery system (uPA-CS/HS@RGD-ODE) was constructed. It is composed of cationic chitosan and anionic heparin assembled in a layer by layer structure, followed by surface modification using RGD peptide and 2-(N-oxide-N,N-diethylamino) ethylmethacrylate (ODE) before encapsulation of urokinase-type plasminogen activator (uPA). The effect of chitosan on the basic performances of uPA-CS/HS@RGD-ODE was estimated. The in vitro results suggest the uPA carrier, CS/HS@RGD-ODE, displayed outstanding targeting specific to activated platelets (61 %) and microenvironment-responsiveness at pH 6.5, facilitating thrombus-targeting and a controlled drug release, respectively. Most importantly, in vivo experiment suggests ODE from uPA-CS/HS@RGD-ODE substantially extends the half-life of uPA (120 min), as uPA-CS/HS@RGD-ODE can adhere onto erythrocytes and deliver uPA under cover of erythrocytes enabling a prolonged circulation time in the bloodstream. Further tail vein and abdominal aorta thrombosis models confirmed uPA-CS/HS@RGD-ODE exhibited superior targeting and thrombolysis capabilities compared to systemic administration of free uPA. To the knowledge of authors, this may be the first study to develop new drug carriers for delivery of thrombolytic drugs under the cover of erythrocytes for extended drug half-lives.


Asunto(s)
Quitosano , Portadores de Fármacos , Eritrocitos , Fibrinolíticos , Trombosis , Activador de Plasminógeno de Tipo Uroquinasa , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Quitosano/química , Quitosano/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Portadores de Fármacos/química , Fibrinolíticos/química , Fibrinolíticos/farmacología , Trombosis/tratamiento farmacológico , Liberación de Fármacos , Terapia Trombolítica/métodos , Heparina/química , Heparina/farmacología , Oligopéptidos/química , Oligopéptidos/farmacología , Humanos , Semivida , Ratones , Sistemas de Liberación de Medicamentos/métodos , Masculino , Polisacáridos/química , Polisacáridos/farmacología
5.
Carbohydr Polym ; 343: 122442, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39174123

RESUMEN

Nanocellulose, a versatile and sustainable nanomaterial derived from cellulose fibers, has attracted considerable attention in various fields due to its unique properties. Similar to dietary fibers, nanocellulose is difficult to digest in the human gastrointestinal tract. The indigestible nanocellulose is fermented by gut microbiota, producing metabolites and potentially exhibiting prebiotic activity in intestinal diseases. Additionally, nanocellulose can serve as a matrix material for probiotic protection and show promising prospects for probiotic delivery. In this review, we summarize the classification of nanocellulose, including cellulose nanocrystals (CNC), cellulose nanofibers (CNF), and bacterial nanocellulose (BNC), highlighting their distinct characteristics and applications. We discuss the metabolism-related characteristics of nanocellulose from oral ingestion to colon fermentation and introduce the prebiotic activity of nanocellulose in intestinal diseases. Furthermore, we provide an overview of commonly used nanocellulose-based encapsulation techniques, such as emulsification, extrusion, freeze drying, and spray drying, as well as the delivery systems employing nanocellulose matrix materials, including microcapsules, emulsions, and hydrogels. Finally, we discuss the challenges associated with nanocellulose metabolism, prebiotic functionality, encapsulation techniques, and delivery systems using nanocellulose matrix material for probiotics. This review will provide new insight into the application of nanocellulose in the treatment of intestinal diseases and probiotic delivery.


Asunto(s)
Celulosa , Microbioma Gastrointestinal , Nanopartículas , Probióticos , Celulosa/química , Celulosa/metabolismo , Humanos , Nanopartículas/química , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Animales , Nanofibras/química , Fermentación , Prebióticos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos
6.
Fluids Barriers CNS ; 21(1): 65, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138578

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disorder with minimally effective treatment options. An important hurdle in ALS drug development is the non-invasive therapeutic access to the motor cortex currently limited by the presence of the blood-brain barrier (BBB). Focused ultrasound and microbubble (FUS+ MB) treatment is an emerging technology that was successfully used in ALS patients to temporarily open the cortical BBB. However, FUS+ MB-mediated drug delivery across ALS patients' BBB has not yet been reported. Similarly, the effects of FUS+ MB on human ALS BBB cells remain unexplored. METHODS: Here we established the first FUS+ MB-compatible, fully-human ALS patient-cell-derived BBB model based on induced brain endothelial-like cells (iBECs) to study anti-TDP-43 antibody delivery and FUS+ MB bioeffects in vitro. RESULTS: Generated ALS iBECs recapitulated disease-specific hallmarks of BBB pathology, including reduced BBB integrity and permeability, and TDP-43 proteinopathy. The results also identified differences between sporadic ALS and familial (C9orf72 expansion carrying) ALS iBECs reflecting patient heterogeneity associated with disease subgroups. Studies in these models revealed successful ALS iBEC monolayer opening in vitro with no adverse cellular effects of FUS+ MB as reflected by lactate dehydrogenase (LDH) release viability assay and the lack of visible monolayer damage or morphology change in FUS+ MB treated cells. This was accompanied by the molecular bioeffects of FUS+ MB in ALS iBECs including changes in expression of tight and adherens junction markers, and drug transporter and inflammatory mediators, with sporadic and C9orf72 ALS iBECs generating transient specific responses. Additionally, we demonstrated an effective increase in the delivery of anti-TDP-43 antibody with FUS+ MB in C9orf72 (2.7-fold) and sporadic (1.9-fold) ALS iBECs providing the first proof-of-concept evidence that FUS+ MB can be used to enhance the permeability of large molecule therapeutics across the BBB in a human ALS in vitro model. CONCLUSIONS: Together, this study describes the first characterisation of cellular and molecular responses of ALS iBECs to FUS+ MB and provides a fully-human platform for FUS+ MB-mediated drug delivery screening on an ALS BBB in vitro model.


Asunto(s)
Esclerosis Amiotrófica Lateral , Barrera Hematoencefálica , Proteínas de Unión al ADN , Microburbujas , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Humanos , Proteínas de Unión al ADN/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/metabolismo , Anticuerpos/administración & dosificación , Ondas Ultrasónicas , Células Cultivadas
7.
Mikrochim Acta ; 191(9): 541, 2024 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-39150483

RESUMEN

The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.


Asunto(s)
Antineoplásicos , Sistemas de Liberación de Medicamentos , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Portadores de Fármacos/química , Animales , Nanomedicina Teranóstica/métodos
8.
AAPS PharmSciTech ; 25(7): 196, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174848

RESUMEN

In this study, N, N '-bis {4- [(α-L- rhamnosyloxy) benzyl]} thiourea (PG-1), a phenolic glycoside compound was purified from Moringa seed. The PG-1 has attracted extensive attention due to its anti-cancer, antioxidant, anti-inflammatory and hypoglycemic properties. However, some of its physicochemical properties such as oral bioavailability has not been studied. Herein, a highly purified PG-1 was extracted and incorporated in multiple layered liposomes (PG-1-L) to avoid its burst release and enhance oral bioavailability. After appropriate characterization, it was discovered that the obtained PG-1-L was stable, homogeneous and well dispersed with the average particle size being 89.26 ± 0.23 nm. Importantly, the in vitro release and in vivo oral bioavailability of PG-1-L were significantly improved compared with PG-1. In addition, MTT results showed that compared with the free PG-1, PG-1-L displayed obvious inhibitory effect on the HepG2 cells, while the inhibitory effect on healthy non-malignant 3T6 and LO-2 cells was not significant, indicating that PG-1-L had high safety. In conclusion, PG-1-L can be used as a promising delivery system and an ideal novel approach to improve the oral bioavailability and anticancer activity of PG-1.


Asunto(s)
Disponibilidad Biológica , Glicósidos , Liposomas , Moringa oleifera , Fenoles , Semillas , Moringa oleifera/química , Semillas/química , Humanos , Glicósidos/química , Glicósidos/administración & dosificación , Glicósidos/farmacología , Glicósidos/aislamiento & purificación , Animales , Células Hep G2 , Fenoles/administración & dosificación , Fenoles/química , Fenoles/aislamiento & purificación , Fenoles/farmacocinética , Tamaño de la Partícula , Sistemas de Liberación de Medicamentos/métodos , Ratones , Masculino , Ratas , Administración Oral , Química Farmacéutica/métodos , Ratas Sprague-Dawley
9.
J Nanobiotechnology ; 22(1): 504, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39175020

RESUMEN

Pyroptosis, a lytic form of cell death mediated by the gasdermin family, is characterized by cell swelling and membrane rupture. Inducing pyroptosis in cancer cells can enhance antitumor immune responses and is a promising strategy for cancer therapy. However, excessive pyroptosis may trigger the development of inflammatory diseases due to immoderate and continuous inflammatory reactions. Nanomaterials and nanobiotechnology, renowned for their unique advantages and diverse structures, have garnered increasing attention owing to their potential to induce pyroptosis in diseases such as cancer. A nano-delivery system for drug-induced pyroptosis in cancer cells can overcome the limitations of small molecules. Furthermore, nanomedicines can directly induce and manipulate pyroptosis. This review summarizes and discusses the latest advancements in nanoparticle-based treatments with pyroptosis among inflammatory diseases and cancer, focusing on their functions and mechanisms and providing valuable insights into selecting nanodrugs for pyroptosis. However, the clinical application of these strategies still faces challenges owing to a limited understanding of nanobiological interactions. Finally, future perspectives on the emerging field of pyroptotic nanomaterials are presented.


Asunto(s)
Inflamación , Nanopartículas , Neoplasias , Piroptosis , Piroptosis/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Animales , Inflamación/tratamiento farmacológico , Nanomedicina/métodos , Sistemas de Liberación de Medicamentos/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
10.
J Nanobiotechnology ; 22(1): 482, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39135039

RESUMEN

Treatment-induced ototoxicity and accompanying hearing loss are a great concern associated with chemotherapeutic or antibiotic drug regimens. Thus, prophylactic cure or early treatment is desirable by local delivery to the inner ear. In this study, we examined a novel way of intratympanically delivered sustained nanoformulation by using crosslinked hybrid nanoparticle (cHy-NPs) in a thermoresponsive hydrogel i.e. thermogel that can potentially provide a safe and effective treatment towards the treatment-induced or drug-induced ototoxicity. The prophylactic treatment of the ototoxicity can be achieved by using two therapeutic molecules, Flunarizine (FL: T-type calcium channel blocker) and Honokiol (HK: antioxidant) co-encapsulated in the same delivery system. Here we investigated, FL and HK as cytoprotective molecules against cisplatin-induced toxic effects in the House Ear Institute - Organ of Corti 1 (HEI-OC1) cells and in vivo assessments on the neuromast hair cell protection in the zebrafish lateral line. We observed that cytotoxic protective effect can be enhanced by using FL and HK in combination and developing a robust drug delivery formulation. Therefore, FL-and HK-loaded crosslinked hybrid nanoparticles (FL-cHy-NPs and HK-cHy-NPs) were synthesized using a quality-by-design approach (QbD) in which design of experiment-central composite design (DoE-CCD) following the standard least-square model was used for nanoformulation optimization. The physicochemical characterization of FL and HK loaded-NPs suggested the successful synthesis of spherical NPs with polydispersity index < 0.3, drugs encapsulation (> 75%), drugs loading (~ 10%), stability (> 2 months) in the neutral solution, and appropriate cryoprotectant selection. We assessed caspase 3/7 apopototic pathway in vitro that showed significantly reduced signals of caspase 3/7 activation after the FL-cHy-NPs and HK-cHy-NPs (alone or in combination) compared to the CisPt. The final formulation i.e. crosslinked-hybrid-nanoparticle-embedded-in-thermogel was developed by incorporating drug-loaded cHy-NPs in poloxamer-407, poloxamer-188, and carbomer-940-based hydrogel. A combination of artificial intelligence (AI)-based qualitative and quantitative image analysis determined the particle size and distribution throughout the visible segment. The developed formulation was able to release the FL and HK for at least a month. Overall, a highly stable nanoformulation was successfully developed for combating treatment-induced or drug-induced ototoxicity via local administration to the inner ear.


Asunto(s)
Nanopartículas , Pez Cebra , Animales , Nanopartículas/química , Oído Interno/efectos de los fármacos , Hidrogeles/química , Cisplatino/farmacología , Cisplatino/química , Línea Celular , Compuestos de Bifenilo/química , Sistemas de Liberación de Medicamentos/métodos , Lignanos/química , Lignanos/farmacología , Lignanos/administración & dosificación , Ratones , Supervivencia Celular/efectos de los fármacos
11.
Int J Nanomedicine ; 19: 8175-8188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39157733

RESUMEN

Exosomes belong to a subgroup of extracellular vesicles secreted by various cells and are involved in intercellular communication and material transfer. In recent years, exosomes have been used as drug delivery carriers because of their natural origin, high stability, low immunogenicity and high engineering ability. However, achieving targeted drug delivery with exosomes remains challenging. In this paper, a phage display technology was used to screen targeted peptides, and different surface modification strategies of targeted peptide exosomes were reviewed. In addition, the application of peptide-targeted exosomes in pulmonary diseases was also summarised.


Asunto(s)
Sistemas de Liberación de Medicamentos , Exosomas , Pulmón , Péptidos , Exosomas/química , Exosomas/metabolismo , Humanos , Péptidos/química , Péptidos/farmacología , Pulmón/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Enfermedades Pulmonares/tratamiento farmacológico , Animales , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Técnicas de Visualización de Superficie Celular/métodos
12.
Int J Nanomedicine ; 19: 8237-8251, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39157735

RESUMEN

Background: Breast cancer presents significant challenges due to the limited effectiveness of available treatments and the high likelihood of recurrence. iRGD possesses both RGD sequence and C-terminal sequence and has dual functions of targeting and membrane penetration. iRGD-modified nanocarriers can enhance drug targeting of tumor vascular endothelial cells and penetration of new microvessels, increasing drug concentration in tumor tissues. Methods: The amidation reaction was carried out between SiO2/AuNCs and iRGD/PTX, yielding a conjugated drug delivery system (SiO2/AuNCs-iRGD/PTX, SAIP@NPs). The assessment encompassed the characterization of the morphology, particle size distribution, physicochemical properties, in vitro release profile, cytotoxicity, and cellular uptake of SAIP@NPs. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed using a small animal in vivo imaging system and a tumor-bearing nude mice model, respectively. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed utilizing a small animal in vivo imaging system and an in situ nude mice breast cancer xenograft model, respectively. Results: The prepared SAIP@NPs exhibited decent stability and a certain slow-release effect in phosphate buffer (PBS, pH 7.4). In vitro studies had shown that, due to the dual functions of transmembrane and targeting of iRGD peptide, SAIP@NPs exhibited strong binding to integrin αvß3, which was highly expressed on the membrane of MDA-MB-231 cells, improving the uptake capacity of tumor cells, inhibiting the rapid growth of tumor cells, and promoting tumor cell apoptosis. The results of animal experiments further proved that SAIP@NPs had longer residence time in tumor sites, stronger anti-tumor effect, and no obvious toxicity to major organs of experimental animals. Conclusion: The engineered SAIP@NPs exhibited superior functionalities including efficient membrane permeability, precise tumor targeting, and imaging, thereby significantly augmenting the therapeutic efficacy against breast cancer with a favorable safety profile.


Asunto(s)
Neoplasias de la Mama , Oro , Nanopartículas del Metal , Ratones Desnudos , Oligopéptidos , Dióxido de Silicio , Animales , Dióxido de Silicio/química , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Humanos , Oligopéptidos/química , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Oro/química , Oro/farmacocinética , Oro/farmacología , Ratones , Línea Celular Tumoral , Nanopartículas del Metal/química , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Paclitaxel/química , Paclitaxel/farmacología , Paclitaxel/farmacocinética , Paclitaxel/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Tamaño de la Partícula , Células MCF-7
13.
Sci Rep ; 14(1): 19228, 2024 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164355

RESUMEN

For successful treatment of diseases, sufficient therapeutics must be provided to the body. Microneedle applications in therapeutic delivery and analytics sampling are restricted because of various issues, including smaller area for drug loading and analytics sampling. To achieve sufficient drug loading and analytics sampling and improve drug penetration while maintaining painless administration, patch-type microneedle arrays were designed and fabricated using polymer casting from a conical cavity mold. Microcavities were formed on a carbon plate via micromechanical machining. A porous polymer layer was coated on a microneedle patch (MNP). The pores of the porous polymer layer provided space and channels for drug delivery. A pH-sensitive polymer layer was employed to cap the porous polymer layer, which prevented drug leakage during storage and provided a stimulus drug release in response to body pH conditions. The drug can be delivered through holes connected to both sides of the patch. The drug release of the MNP was investigated in vitro and in vivo and showed conceptual proof that these MNs have the potential to enhance treatment protocols for various diseases with the flexibility of coating and therapeutic materials and offer significant scope for further variations and advancement.


Asunto(s)
Carbono , Sistemas de Liberación de Medicamentos , Agujas , Sistemas de Liberación de Medicamentos/métodos , Carbono/química , Animales , Liberación de Fármacos , Microinyecciones/instrumentación , Microinyecciones/métodos , Porosidad , Concentración de Iones de Hidrógeno , Polímeros/química , Ratones
14.
J Nanobiotechnology ; 22(1): 498, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164657

RESUMEN

Microcarrier is a promising drug delivery system demonstrating significant value in treating cancers. One of the main goals is to devise microcarriers with ingenious structures and functions to achieve better therapeutic efficacy in tumors. Here, inspired by the nucleus-cytoplasm structure of cells and the material exchange reaction between them, we develop a type of biorthogonal compartmental microparticles (BCMs) from microfluidics that can separately load and sequentially release cyclooctene-modified doxorubicin prodrug (TCO-DOX) and tetrazine-modified indocyanine green (Tz-ICG) for tumor therapy. The Tz-ICG works not only as an activator for TCO-DOX but also as a photothermal agent, allowing for the combination of bioorthogonal chemotherapy and photothermal therapy (PTT). Besides, the modification of DOX with cyclooctene significantly decreases the systemic toxicity of DOX. As a result, the developed BCMs demonstrate efficient in vitro tumor cell eradication and exhibit notable tumor growth inhibition with favorable safety. These findings illustrate that the formulated BCMs establish a platform for bioorthogonal prodrug activation and localized delivery, holding significant potential for cancer therapy and related applications.


Asunto(s)
Doxorrubicina , Sistemas de Liberación de Medicamentos , Verde de Indocianina , Terapia Fototérmica , Profármacos , Doxorrubicina/farmacología , Doxorrubicina/química , Terapia Fototérmica/métodos , Humanos , Profármacos/farmacología , Profármacos/química , Animales , Verde de Indocianina/química , Verde de Indocianina/farmacología , Ratones , Sistemas de Liberación de Medicamentos/métodos , Línea Celular Tumoral , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Ciclooctanos/química , Ciclooctanos/farmacología , Ratones Endogámicos BALB C , Antineoplásicos/farmacología , Antineoplásicos/química , Femenino
15.
J Nanobiotechnology ; 22(1): 499, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164747

RESUMEN

Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.


Asunto(s)
Barrera Hematoencefálica , Sistemas de Liberación de Medicamentos , Accidente Cerebrovascular Isquémico , Nanopartículas , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Estrés Oxidativo/efectos de los fármacos , Sistema de Administración de Fármacos con Nanopartículas/química , Isquemia Encefálica/tratamiento farmacológico , Nanomedicina/métodos , Encéfalo/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/química
16.
Drug Deliv ; 31(1): 2392755, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39166341

RESUMEN

BACKGROUND: There exists an unfulfilled requirement for effective cochlear pharmacotherapy. Controlled local drug delivery could lead to effective bioavailability. The round window niche (RWN), a cavity in the middle ear, is connected to the cochlea via a membrane through which drug can diffuse. We are developing individualized drug-eluting RWN implants (RNIs). To test their effectiveness in guinea pigs, a commonly used model in cochlear pharmacology studies, it is first necessary to develop guinea pig RNIs (GP-RNI). METHODS: Since guinea pigs do not have a RWN such as it is present in humans and to reduce the variables in in vivo studies, a one-size-fits-all GP-RNI model was designed using 12 data sets of Dunkin-Hartley guinea pigs. The model was 3D-printed using silicone. The accuracy and precision of printing, distribution of the sample ingredient dexamethasone (DEX), biocompatibility, bio-efficacy, implantability and drug release were tested in vitro. The GP-RNI efficacy was validated in cochlear implant-traumatized guinea pigs in vivo. RESULTS: The 3D-printed GP-RNI was precise, accurate and fitted in all tested guinea pig RWNs. DEX was homogeneously included in the silicone. The GP-RNI containing 1% DEX was biocompatible, bio-effective and showed a two-phase and sustained DEX release in vitro, while it reduced fibrous tissue growth around the cochlear implant in vivo. CONCLUSIONS: We developed a GP-RNI that can be used for precise inner ear drug delivery in guinea pigs, providing a reliable platform for testing the RNI's safety and efficacy, with potential implications for future clinical translation.


Asunto(s)
Implantes Cocleares , Dexametasona , Sistemas de Liberación de Medicamentos , Ventana Redonda , Cobayas , Animales , Ventana Redonda/efectos de los fármacos , Ventana Redonda/metabolismo , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Dexametasona/farmacología , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Impresión Tridimensional , Cóclea/efectos de los fármacos
17.
Int J Nanomedicine ; 19: 8337-8352, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161359

RESUMEN

Osteoarthritis (OA) is a degenerative disease commonly seen in middle-aged and elderly people. Multiple cytokines are involved in the local tissue damage in OA. Currently, non-pharmacologic and surgical interventions are the main conventional approaches for the treatment of OA. In terms of pharmaceutical drug therapy, NSAIDs and acetaminophen are mainly used to treat OA. However, it is prone to various adverse reactions such as digestive tract ulcer, thromboembolism, prosthesis loosening, nerve injury and so on. With the in-depth study of OA, more and more novel topical drug delivery strategies and vehicles have been developed, which can make up for the shortcomings of traditional dosage forms, improve the bioavailability of drugs, and significantly reduce drug side effects. This review summarizes the immunopathogenesis, treatment guidelines, and progress and challenges of topical delivery technologies of OA, with some perspectives on the future pharmacological treatment of OA proposed.


Asunto(s)
Antiinflamatorios no Esteroideos , Sistemas de Liberación de Medicamentos , Osteoartritis , Humanos , Osteoartritis/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Antiinflamatorios no Esteroideos/administración & dosificación , Administración Tópica , Acetaminofén/administración & dosificación , Animales , Disponibilidad Biológica
18.
Int J Nanomedicine ; 19: 8353-8371, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161357

RESUMEN

Introduction: Overactive bladder (OAB) is a highly prevalent condition with limited treatment options due to poor efficacy, side effects, and patient compliance. Novel drug delivery systems that can target the bladder wall may improve OAB therapy. Methods: We explored a polydopamine (PDA)-coated lactobacillus platform as a potential carrier for localized OAB treatment. Urinary microbiome profiling was performed to identify the presence of lactobacillus in healthy and OAB groups. Lactobacillus-PDA nanoparticles were synthesized and characterized by electron microscopy and spectrophotometry. A rat bladder perfusion model and human bladder smooth muscle cell spheroids were used to assess the distribution and penetration of the nanoparticles. The efficacy of the Lactobacillus-PDA system (LPS) for delivering the antimuscarinic drug solifenacin was evaluated in an OAB rat model. Results: Urinary microbiome profiling revealed lactobacillus as a dominant genus in both healthy and OAB groups. The synthesized Lactobacillus-PDA nanoparticles exhibited uniform size and optical properties. In the rat bladder perfusion model, the nanoparticles distributed throughout the bladder wall and smooth muscle without toxicity. The nanoparticles also penetrated human bladder smooth muscle cell spheroids. In the OAB rat model, LPS facilitated the delivery of solifenacin and improved treatment efficacy. Discussion: The results highlight LPS as a promising drug carrier for targeted OAB therapy via penetration into bladder tissues. This bacteriotherapy approach may overcome limitations of current systemic OAB medications. Lactobacillus, a probiotic bacterium present in the urinary tract microbiome, was hypothesized to adhere to and penetrate the bladder wall when coated with PDA nanoparticles, making it a suitable candidate for localized drug delivery.


Asunto(s)
Indoles , Lactobacillus , Microbiota , Polímeros , Vejiga Urinaria Hiperactiva , Vejiga Urinaria , Animales , Indoles/química , Indoles/farmacocinética , Vejiga Urinaria Hiperactiva/terapia , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Humanos , Polímeros/química , Microbiota/efectos de los fármacos , Ratas , Esferoides Celulares , Succinato de Solifenacina/farmacocinética , Succinato de Solifenacina/química , Succinato de Solifenacina/administración & dosificación , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos , Femenino , Miocitos del Músculo Liso/efectos de los fármacos , Antagonistas Muscarínicos/farmacocinética , Antagonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/química , Antagonistas Muscarínicos/administración & dosificación , Portadores de Fármacos/química
19.
Int J Nanomedicine ; 19: 8091-8113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161361

RESUMEN

The current treatments for wound healing still exhibit drawbacks due to limited availability at the action sites, susceptibility to degradation, and immediate drug release, all of which are detrimental in chronic conditions. Nano-modification strategies, offering various advantages that can enhance the physicochemical properties of drugs, have been employed in efforts to maximize the efficacy of wound healing medications. Nowadays, nanostructured lipid carriers (NLCs) provide drug delivery capabilities that can safeguard active compounds from environmental influences and enable controlled release profiles. Consequently, NLCs are considered an alternative therapy to address the challenges encountered in wound treatment. This review delves into the application of NLCs in drug delivery for wound healing, encompassing discussions on their composition, preparation methods, and their impact on treatment effectiveness. The modification of drugs into the NLC model can be facilitated using relatively straightforward technologies such as pressure-based processes, emulsification techniques, solvent utilization methods, or phase inversion. Moreover, NLC production with minimal material compositions can accommodate both single and combination drug delivery. Through in vitro, in vivo, and clinical studies, it has been substantiated that NLCs can enhance the therapeutic potential of various drug types in wound healing treatments. NLCs enhance efficacy by reducing the active substance particle size, increasing solubility and bioavailability, and prolonging drug release, ensuring sustained dosage at the wound site for chronic wounds. In summary, NLCs represent an effective nanocarrier system for optimizing the bioavailability of active pharmacological ingredients in the context of wound healing.


Asunto(s)
Portadores de Fármacos , Lípidos , Nanoestructuras , Cicatrización de Heridas , Cicatrización de Heridas/efectos de los fármacos , Humanos , Portadores de Fármacos/química , Lípidos/química , Nanoestructuras/química , Animales , Tamaño de la Partícula , Liberación de Fármacos , Sistemas de Liberación de Medicamentos/métodos , Preparaciones de Acción Retardada/química , Disponibilidad Biológica
20.
Int J Nanomedicine ; 19: 8373-8400, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161363

RESUMEN

Despite all major advancements in drug discovery and development in the pharmaceutical industry, cancer is still one of the most arduous challenges for the scientific community. The implications of nanotechnology have certainly resolved major issues related to conventional anticancer modalities; however, the undesired recognition of nanoparticles (NPs) by the mononuclear phagocyte system (MPS), their poor stability in biological fluids, premature release of payload, and low biocompatibility have restricted their clinical translation. In recent decades, chitosan (CS)-based nanodelivery systems (eg, polymeric NPs, micelles, liposomes, dendrimers, conjugates, solid lipid nanoparticles, etc.) have attained promising recognition from researchers for improving the pharmacokinetics and pharmacodynamics of chemotherapeutics. However, the specialty of this review is to mainly focus on and critically discuss the targeting potential of various CS-based NPs for treatment of different types of cancer. Based on their delivery mechanisms, we classified CS-based NPs into stimuli-responsive, passive, or active targeting nanosystems. Moreover, various functionalization strategies (eg, grafting with polyethylene glycol (PEG), hydrophobic substitution, tethering of stimuli-responsive linkers, and conjugation of targeting ligands) adapted to the architecture of CS-NPs for target-specific delivery of chemotherapeutics have also been considered. Nevertheless, CS-NPs based therapeutics hold great promise for improving therapeutic outcomes while mitigating the off-target effects of chemotherapeutics, a long-term safety profile and clinical testing in humans are warranted for their successful clinical translation.


Asunto(s)
Antineoplásicos , Quitosano , Nanopartículas , Neoplasias , Humanos , Quitosano/química , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Animales , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Nanomedicina/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...