Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502155

RESUMEN

The present study aims to investigate the loco-regional tolerability and injection parameters (i.e., flow rate and administration volume) of an in situ forming depot (ISFD) in Göttingen minipigs, to secure both the therapeutic procedure and compliance in chronic medical prescriptions. The ISFD BEPO® technology (MedinCell S.A.) is investigated over 10 days, after a single subcutaneous injection of test item based on a DMSO solution of diblock and triblock polyethylene glycol-polylactic acid copolymers. Injection sites are systematically observed for macroscopic loco-regional skin reactions as well as ultrasound scanning, enabling longitudinal in vivo imaging of the depot. Observations are complemented by histopathological examinations at 72 h and 240 h post-injection. Overall, no treatment-emergent adverse effects are macroscopically or microscopically observed at the subcutaneous injection sites, for the tested injection flow rates of 1 and 8 mL/min and volumes of 0.2 and 1 mL. The histopathology examination confirms an expected foreign body reaction, with an intensity depending on the injected volume. The depot morphology is similar irrespective of the administration flow rates. These results indicate that the ISFD BEPO® technology can be considered safe when administered subcutaneously in Göttingen minipigs, a human-relevant animal model for subcutaneous administrations, in the tested ranges.


Asunto(s)
Vías de Administración de Medicamentos/veterinaria , Inyecciones Subcutáneas/efectos adversos , Inyecciones Subcutáneas/métodos , Animales , Inmunohistoquímica , Piel/diagnóstico por imagen , Piel/efectos de los fármacos , Piel/patología , Porcinos , Porcinos Enanos , Ultrasonografía
2.
Molecules ; 26(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800940

RESUMEN

This article presents the evaluation of diblock and triblock poly(ethylene glycol)-b-poly(1,3-trimethylene carbonate) amphiphilic copolymers (PEG-PTMCs) as excipients for the formulation of long-acting injectables (LAIs). Copolymers were successfully synthesised through bulk ring-opening polymerisation. The concomitant formation of PTMC homopolymer could not be avoided irrespective of the catalyst amount, but the by-product could easily be removed by gel chromatography. Pure PEG-PTMCs undergo faster erosion in vivo than their corresponding homopolymer. Furthermore, these copolymers show outstanding stability compared to their polyester analogues when formulated with amine-containing reactive drugs, which makes them particularly suitable as LAIs for the sustained release of drugs susceptible to acylation.


Asunto(s)
Materiales Biocompatibles/metabolismo , Dioxanos/química , Polietilenglicoles/química , Polímeros/química , Polímeros/metabolismo , Acilación , Animales , Materiales Biocompatibles/administración & dosificación , Masculino , Polímeros/administración & dosificación , Ratas , Ratas Sprague-Dawley
3.
ACS Appl Mater Interfaces ; 15(48): 55495-55509, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38011651

RESUMEN

This paper presents the in vitro and in vivo degradation of BEPO, a marketed in situ forming depot technology used for the formulation of long-acting injectables. BEPO is composed of a solution of a blend of poly(ethylene glycol)-block-poly(lactic acid) (PEG-PLA) triblock and diblock in an organic solvent, where a therapeutic agent may be dissolved or suspended. Upon contact with an aqueous environment, the solvent diffuses and the polymers precipitate, entrapping the drug and forming a reservoir. Two representative BEPO compositions were subjected to a 3-month degradation study in vitro by immersion in phosphate-buffered saline at 37 °C and in vivo after subcutaneous injection in minipig. The material erosion rate, as a surrogate of the bioresorption, determined via the depot weight loss, changed substantially, depending on the composition and content of polymers within the test item. The swelling properties and internal morphology of depots were shown to be highly dependent on the solvent exchange rate during the precipitation step. Thermal analyses displayed an increase of the depot glass transition temperature over the degradation process, with no crystallinity observed at any stage. The chemical composition of degraded depots was determined by 1H NMR and gel permeation chromatography and demonstrated an enrichment in homopolymers, i.e., free PLA and (m)PEG, to the detriment of (m)PEG-PLA copolymers in both formulations. It was observed that the relative ratio of the degradants within the depot is driven by the initial polymer composition. Interestingly, in vitro and in vivo results showed very good qualitative consistency. Taken together, the outcomes from this study demonstrate that the different hydrolytic degradation behaviors of the BEPO compositions can be tuned by adjusting the polymer composition of the formulation.


Asunto(s)
Polietilenglicoles , Polímeros , Animales , Porcinos , Porcinos Enanos , Polímeros/química , Polietilenglicoles/química , Sistemas de Liberación de Medicamentos , Poliésteres/química , Solventes/química
4.
Expert Opin Drug Deliv ; 19(8): 927-944, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35899474

RESUMEN

INTRODUCTION: The development of long-acting injectables (LAIs) for protein and peptide therapeutics has been a key challenge over the last 20 years. If these molecules offer advantages due to their high specificity and selectivity, their controlled release may confer several additional benefits in terms of extended half-life, local delivery, and patient compliance. AREA COVERED: This manuscript aims to give an overview of peptide and protein-based LAIs from an industrial perspective, describing both approved and promising technologies (with exceptions of protein engineering strategies and devices), their advantages and potential improvements to aid their access to the market. EXPERT OPINION: Many LAIs have been developed for peptides, with formulations on the market for several decades. On the contrary, LAIs for proteins are still far from the market and issues related to manufacturing and sterilization of these products still need to be overcome. In situ forming depots (ISFDs), whose simple manufacturing conditions and easy administration procedures (without reconstitution) are strong advantages, appear as one of the most promising technologies for the delivery of these molecules. In this regard, the approval of ELIGARD® in the early 2000's (which still requires a complex reconstitution process), paved the way for the development of second-generation, ready-to-use ISFD technologies like BEPO® and FluidCrystal®.


Asunto(s)
Péptidos , Proteínas , Preparaciones de Acción Retardada , Humanos , Inyecciones
5.
J Control Release ; 341: 578-590, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34915070

RESUMEN

Monoclonal antibodies (mAbs) are large size molecules that have demonstrated high therapeutic potential for the treatment of cancer or autoimmune diseases. Despite some excellent results, their intravenous administration results in high plasma concentration. This triggers off-target effects and sometimes poor targeted tissue distribution. To circumvent this issue, we investigated a local controlled-delivery approach using an in situ forming depot technology. Two clinically relevant mAbs, rituximab (RTX) and daratumumab (DARA), were formulated using an injectable technology based on biodegradable PEG-PLA copolymers. The stability and controlled release features of the formulations were investigated. HPLC and mass spectrometry revealed the preservation of the protein structure. In vitro binding of formulated antibodies to their target antigens and to their cellular FcγRIIIa natural killer cell receptor was fully maintained. Furthermore, encapsulated RTX was as efficient as classical intravenous RTX treatment to inhibit the in vivo tumor growth of malignant human B cells in immunodeficient NSG mice. Finally, the intra-articular administration of the formulated mAbs yielded a sustained local release associated with a lower plasma concentration compared to the intra-articular delivery of non-encapsulated mAbs. Our results demonstrate that the utilization of this polymeric technology is a reliable alternative for the local delivery of fully functional clinically relevant mAbs.


Asunto(s)
Polímeros , Animales , Preparaciones de Acción Retardada/química , Ratones , Polímeros/química
6.
Pharmaceutics ; 13(5)2021 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922166

RESUMEN

This article describes the utilization of (methoxy)poly(ethylene glycol)-b-poly(1,3-trimethylene carbonate) ((m)PEG-PTMC) diblock and triblock copolymers for the formulation of in situ forming depot long-acting injectables by solvent exchange. The results shown in this manuscript demonstrate that it is possible to achieve long-term drug deliveries from suspension formulations prepared with these copolymers, with release durations up to several months in vitro. The utilization of copolymers with different PEG and PTMC molecular weights affords to modulate the release profile and duration. A pharmacokinetic study in rats with meloxicam confirmed the feasibility of achieving at least 28 days of sustained delivery by using this technology while showing good local tolerability in the subcutaneous environment. The characterization of the depots at the end of the in vivo study suggests that the rapid phase exchange upon administration and the surface erosion of the resulting depots are driving the delivery kinetics from suspension formulations. Due to the widely accepted utilization of meloxicam as an analgesic drug for animal care, the results shown in this article are of special interest for the development of veterinary products aiming at a very long-term sustained delivery of this therapeutic molecule.

7.
J Control Release ; 336: 457-468, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34214596

RESUMEN

The generation of acylated impurities has represented an important hurdle in the development of long acting injectables for therapeutic peptides using biocompatible polymers with a polyester moiety. We investigated here an in situ forming depot (ISFD) technology that uses polyethylene glycol - polyester copolymers and a solvent exchange mechanism to promote depot formation. This technology has shown promise in formulating small molecules as well as therapeutic proteins. In the present work, using the well-known somatostatin analog octreotide acetate (OctAc) as a model molecule, we evaluated this delivery platform to release therapeutic peptides. Peptide acylation was found to be pronounced in the formulation, while it was very limited once the depot was formed and during the release process. The octreotide acylation pattern was fully characterized by LC-MS/MS. Moreover, it was demonstrated that exchanging the acetate anion with more hydrophobic counterions like pamoate or lauryl sulfate allowed to greatly improve the peptide stability profile, as well as the formulation release performance. Finally, the in vivo evaluation through pharmacokinetics studies in rat of these new octreotide salts in ISFD formulations showed that octreotide was quantifiable up to four weeks post-administration with a high bioavailability and an acceptable initial burst.


Asunto(s)
Octreótido , Espectrometría de Masas en Tándem , Animales , Cromatografía Liquida , Cinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Tecnología
8.
Chemistry ; 16(35): 10879-86, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20661959

RESUMEN

Mesoporous bioactive glasses (MBGs) associated with an anti-osteoporotic drug (ipriflavone) have been prepared. With this aim, MBGs were functionalised with different organic groups by following a post-grafting method, thus retaining the mesoporous network of the bioactive substrates. Drug-delivery tests were carried out by using ipriflavone as a hydrophobic model drug. Our results revealed that by means of the proper functionalisation, most of the drug is retained in the mesoporous network. By tailoring the hydrophobicity of the surface with functional groups, the drug-material link can be tuned, thereby ensuring the long-term delivery of ipriflavone. In vitro bioactive tests demonstrate that these systems exhibit the same excellent behaviour of non-functionalised MBGs. The possibility to add a bone resorption inhibitor such as ipriflavone to highly bioactive materials confirms functionalised MBGs as very promising bone-tissue regeneration systems.


Asunto(s)
Materiales Biocompatibles/química , Cerámica/química , Sistemas de Liberación de Medicamentos/métodos , Vidrio/química , Isoflavonas/química , Regeneración Ósea , Interacciones Hidrofóbicas e Hidrofílicas , Isoflavonas/farmacología
9.
J Control Release ; 319: 416-427, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-31931049

RESUMEN

This article presents BEPO®, an in situ forming depot (ISFD) technology mediated by a solvent-exchange mechanism. The matrix of the in situ formed drug delivery depot is composed of the combination of a diblock (DB) and a triblock (TB) polyethylene glycol-polyester copolymer. This combination offers a broad capability to tune the release of a wide variety of drugs to the desired pharmacokinetics. The work described in the present article demonstrates that the delivery rate and profile can be adjusted by changing the composition of either TB or DB or the relative ratio between them, among other parameters. It has been shown that the polymeric composition of the formulation has a substantial impact on the solvent exchange rate between the organic solvent and the surrounding aqueous medium which subsequently determines the internal structure of the resulting depot and the delivery of the therapeutic cargo. This has been demonstrated studying the in vitro release of two model molecules: bupivacaine and ivermectin. Formulations releasing these drugs have been administered to animal models to show the possibility of delivering therapeutics from weeks to months by using BEPO® technology.


Asunto(s)
Implantes Absorbibles , Polietilenglicoles , Animales , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Cinética , Micelas , Poliésteres
10.
J Tissue Eng Regen Med ; 11(4): 1097-1109, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-25783558

RESUMEN

A major limitation with current tissue-engineering approaches is creating functionally vascularized constructs that can successfully integrate with the host; this often leads to implant failure, due to avascular necrosis. In order to overcome this, the objective of the present work was to develop a method to incorporate growth factor-eluting alginate microparticles (MPs) into freeze-dried, collagen-based scaffolds. A collagen-hydroxyapatite (CHA) scaffold, previously optimized for bone regeneration, was functionalized for the sustained delivery of an angiogenic growth factor, vascular endothelial growth factor (VEGF), with the aim of facilitating angiogenesis and enhancing bone regeneration. VEGF was initially encapsulated in alginate MPs by spray-drying, producing particles of < 10 µm in diameter. This process was found to effectively encapsulate and control VEGF release while maintaining its stability and bioactivity post-processing. These VEGF-MPs were then incorporated into CHA scaffolds, leading to homogeneous distribution throughout the interconnected scaffold pore structure. The scaffolds were capable of sustained release of bioactive VEGF for up to 35 days, which was proficient at increasing tubule formation by endothelial cells in vitro. When implanted in vivo in a rat calvarial defect model, this scaffold enhanced vessel formation, resulting in increased bone regeneration compared to empty-defect and VEGF-free scaffolds. This biologically functionalized scaffold, composed entirely of natural-based materials, may offer an ideal platform to promote angiogenesis and tissue regeneration. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Alginatos/química , Regeneración Ósea/efectos de los fármacos , Colágeno/química , Durapatita/química , Microesferas , Neovascularización Fisiológica/efectos de los fármacos , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Materiales Biocompatibles/farmacología , Preparaciones de Acción Retardada/farmacología , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Masculino , Osteogénesis/efectos de los fármacos , Porosidad , Ratas Wistar , Microtomografía por Rayos X
11.
Acta Biomater ; 48: 110-119, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27773752

RESUMEN

Lysolipid-based thermosensitive liposomes (LTSL) embedded in a chitosan-based thermoresponsive hydrogel matrix (denoted Lipogel) represents a novel approach for the spatiotemporal release of therapeutic agents. The entrapment of drug-loaded liposomes in an injectable hydrogel permits local liposome retention, thus providing a prolonged release in target tissues. Moreover, release can be controlled through the use of a minimally invasive external hyperthermic stimulus. Temporal control of release is particularly important for complex multi-step physiological processes, such as angiogenesis, in which different signals are required at different times in order to produce a robust vasculature. In the present work, we demonstrate the ability of Lipogel to provide a flexible, easily modifiable release platform. It is possible to tune the release kinetics of different drugs providing a passive release of one therapeutic agent loaded within the gel and activating the release of a second LTSL encapsulated agent via a hyperthermic stimulus. In addition, it was possible to modify the drug dosage within Lipogel by varying the duration of hyperthermia. This can allow for adaption of drug dosing in real time. As an in vitro proof of concept with this system, we investigated Lipogels ability to recruit stem cells and then elevate their production of vascular endothelial growth factor (VEGF) by controlling the release of a pro-angiogenic drug, desferroxamine (DFO) with an external hyperthermic stimulus. Initial cell recruitment was accomplished by the passive release of hepatocyte growth factor (HGF) from the hydrogel, inducing a migratory response in cells, followed by the delayed release of DFO from thermosensitive liposomes, resulting in a significant increase in VEGF expression. This delayed release could be controlled up to 14days. Moreover, by changing the duration of the hyperthermic pulse, a fine control over the amount of DFO released was achieved. The ability to trigger the release of therapeutic agents at a specific timepoint and control dosing level through changes in duration of hyperthermia enables sequential multi-dose profiles. STATEMENT OF SIGNIFICANCE: This paper details the development of a heat responsive liposome loaded hydrogel for the controlled release of pro-angiogenic therapeutics. Lysolipid-based thermosensitive liposomes (LTSLs) embedded in a chitosan-based thermoresponsive hydrogel matrix represents a novel approach for the spatiotemporal release of therapeutic agents. This hydrogel platform demonstrates remarkable flexibility in terms of drug scheduling and sequencing, enabling the release of multiple agents and the ability to control drug dosing in a minimally invasive fashion. The possibility to tune the release kinetics of different drugs independently represents an innovative platform to utilise for a variety of treatments. This approach allows a significant degree of flexibility in achieving a desired release profile via a minimally invasive stimulus, enabling treatments to be tuned in response to changing symptoms and complications.


Asunto(s)
Deferoxamina/farmacología , Liberación de Fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Materiales Biocompatibles/farmacología , Movimiento Celular/efectos de los fármacos , Quitosano/química , Glicerofosfatos/química , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Hipertermia Inducida , Liposomas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Adv Healthc Mater ; 4(17): 2649-56, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26414944

RESUMEN

Defects within bones caused by trauma and other pathological complications may often require the use of a range of therapeutics to facilitate tissue regeneration. A number of approaches have been widely utilized for the delivery of such therapeutics via physical encapsulation or chemical immobilization suggesting significant promise in the healing of bone defects. The study focuses on the chemical immobilization of osteostatin, a pentapeptide of the parathyroid hormone (PTHrP107-111), within a collagen-hydroxyapatite scaffold. The chemical attachment method via crosslinking supports as little as 4% release of the peptide from the scaffolds after 21 d whereas non-crosslinking leads to 100% of the peptide being released by as early as 4 d. In vitro characterization demonstrates that this cross-linking method of immobilization supports a pro-osteogenic effect on osteoblasts. Most importantly, when implanted in a critical-sized calvarial defect within a rat, these scaffolds promote significantly greater new bone volume and area compared to nonfunctionalized scaffolds (**p < 0.01) and an empty defect control (***p < 0.001). Collectively, this study suggests that such an approach of chemical immobilization offers greater spatiotemporal control over growth factors and can significantly modulate tissue regeneration. Such a system may be adopted for a range of different proteins and thus offers the potential for the treatment of various complex pathologies that require localized mediation of drug delivery.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Colágeno/química , Durapatita/química , Proteína Relacionada con la Hormona Paratiroidea/química , Fragmentos de Péptidos/química , Andamios del Tejido/química , Animales , Huesos/efectos de los fármacos , Línea Celular , Colágeno/farmacología , Durapatita/farmacología , Masculino , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/química , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Cicatrización de Heridas/efectos de los fármacos
13.
J Control Release ; 207: 112-9, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-25817394

RESUMEN

The clinical utilization of recombinant human bone morphogenetic protein 2 (rhBMP-2) delivery systems for bone regeneration has been associated with very severe side effects, which are due to the non-controlled and non-targeted delivery of the growth factor from its collagen sponge carrier post-implantation which necessitates supraphysiological doses. However, rhBMP-2 presents outstanding regenerative properties and thus there is an unmet need for a biocompatible, fully resorbable delivery system for the controlled, targeted release of this protein. With this in mind, the purpose of this work was to design and develop a delivery system to release low rhBMP-2 doses from a collagen-hydroxyapatite (CHA) scaffold which had previously been optimized for bone regeneration and recently demonstrated significant healing in vivo. In order to enhance the potential for clinical translation by minimizing the design complexity and thus upscaling and regulatory hurdles of the device, a microparticle and chemical functionalization-free approach was chosen to fulfill this aim. RhBMP-2 was combined with a CHA scaffold using a lyophilization fabrication process to produce a highly porous CHA scaffold supporting the controlled release of the protein over the course of 21days while maintaining in vitro bioactivity as demonstrated by enhanced alkaline phosphatase activity and calcium production by preosteoblasts cultured on the scaffold. When implanted in vivo, these materials demonstrated increased levels of healing of critical-sized rat calvarial defects 8weeks post-implantation compared to an empty defect and unloaded CHA scaffold, without eliciting bone anomalies or adjacent bone resorption. These results demonstrate that it is possible to achieve bone regeneration using 30 times less rhBMP-2 than INFUSE®, the current clinical gold standard; thus, this work represents the first step of the development of a rhBMP-2 eluting material with immense clinical potential.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Colágeno/química , Portadores de Fármacos , Hidroxiapatitas/metabolismo , Osteoblastos/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Cráneo/efectos de los fármacos , Andamios del Tejido , Células 3T3 , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2/química , Calcio/metabolismo , Química Farmacéutica , Preparaciones de Acción Retardada , Humanos , Masculino , Ratones , Osteoblastos/metabolismo , Porosidad , Ratas Wistar , Proteínas Recombinantes/química , Cráneo/diagnóstico por imagen , Cráneo/metabolismo , Cráneo/fisiopatología , Factores de Tiempo , Microtomografía por Rayos X
14.
J Control Release ; 198: 71-9, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25481441

RESUMEN

The spatiotemporally controlled delivery of the pro-osteogenic factor rhBMP-2 would overcome most of the severe secondary effects linked to the products delivering this protein for bone regeneration. With this in mind, the aim of the present work was to develop a controlled rhBMP-2 release system using collagen-hydroxyapatite (CHA) scaffolds, which had been previously optimized for bone regeneration, as delivery platforms to produce a device with enhanced capacity for bone repair. Spray-drying and emulsion techniques were used to encapsulate bioactive rhBMP-2 in alginate and PLGA microparticles, with a high encapsulation efficiency. After incorporation of these microparticles into the scaffolds, rhBMP-2 was delivered in a sustained fashion for up to 28days. When tested in vitro with osteoblasts, these eluting materials showed an enhanced pro-osteogenic effect. From these results, an optimal rhBMP-2 eluting scaffold composition was selected and implanted in critical-sized calvarial defects in a rat model, where it demonstrated an excellent healing capacity in vivo. These platforms have an immense potential in the field of tissue regeneration; by tuning the specific therapeutic molecule to the tissue of interest and by utilizing different collagen-based scaffolds, similar systems can be developed for enhancing the healing of a diverse range of tissues and organs.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Colágeno/química , Portadores de Fármacos/administración & dosificación , Durapatita/química , Andamios del Tejido , Alginatos/química , Animales , Proteína Morfogenética Ósea 2/química , Huesos , Línea Celular , Portadores de Fármacos/química , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Ácido Láctico/química , Masculino , Ratones , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Ingeniería de Tejidos
15.
J Control Release ; 187: 158-66, 2014 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-24878185

RESUMEN

Collagen is one of the most attractive materials for the development of matrices for tissue engineering, due to its excellent biocompatibility and non-toxic bioresorption. The present work describes a collagen-based externally controlled drug-eluting scaffold which consists of drug encapsulated thermoresponsive liposomes covalently attached to the surface of a functionalized collagen-based scaffold. The model drug used in this work was PTHrP 107-111, a pentapeptide with pro-osteogenic and antiosteoclastic activity. An osteoconductive collagen-hydroxyapatite scaffold, designed specifically for bone repair, was used as a model scaffold. The results demonstrate that it is possible to modify the kinetics of release of the drug from the scaffold with the application of an external thermal stimulus (42°C, 20min). In vitro studies carried out with pre-osteoblastic MC3T3-E1 cells demonstrated that neither the attachment of liposomes to the surface of the scaffolds nor the hyperthermic pulse negatively affected the ability of cells to attach and proliferate on the scaffolds. Importantly, the on-demand release of PTHrP 107-111 had a pro-osteogenic effect, as shown by the enhancement of alkaline phosphatase activity, an early osteogenic marker, which correlated with increased expression of the osteogenic genes osteopontin and osteocalcin. In conclusion, the scaffold-based release system developed in this study has immense potential for tuning the delivery of a diverse range of drugs which can be applied for the regeneration of a variety of tissue types.


Asunto(s)
Colágeno/química , Osteogénesis/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Andamios del Tejido , Animales , Línea Celular , Durapatita/química , Calor , Liposomas , Ratones
16.
Adv Healthc Mater ; 3(6): 854-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24436226

RESUMEN

A novel drug delivery system, enabling an in situ, thermally triggered drug release is described, consisting of an injectable thermoresponsive chitosan hydrogel containing doxorubicin-loaded thermosensitive liposomes. The design, fabrication, characterization, and an assessment of in vitro bioactivity of this formulation is detailed. Combining on-demand drug delivery with in situ gelation results in a promising candidate for local chemotherapy.


Asunto(s)
Hidrogel de Polietilenoglicol-Dimetacrilato/química , Liposomas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quitosano/química , Doxorrubicina/química , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Glicerofosfatos/química , Humanos , Temperatura
17.
J Biomed Mater Res A ; 89(1): 215-23, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18431760

RESUMEN

The bacterial adherence onto different multifunctional silica-based bioceramics has been evaluated. Staphylococcus aureus and Staphylococcus epidermidis were chosen, as they cause the majority of the implant-related infections in this field. Two SiO2 mesoporous materials (MCM-41, SBA-15), an ordered SiO2-CaO-P2O5 mesoporous glass (OMG), and a biphasic magnetic bioceramic (BMB), were incubated with S. aureus and S. epidermidis for 90 min, and subsequently sonicated to quantify the number of adhered bacteria on each material. It was found that S. aureus and S. epidermidis (10(8) CFU/mL) adhered significantly less to BMB samples when compared to MCM-41, SBA-15, or OMG. However, when the material pores accessible for bacteria in each material were taken into account, the lowest bacterial adherence was found in MCM-41, and the highest in SBA-15. The results show that bacterial adherence is higher on mesoporous bioceramics, although this higher microbial attachment is mainly due to the intergranular porosity and grain size morphology rather than to the mesoporous structure.


Asunto(s)
Adhesión Bacteriana/fisiología , Materiales Biocompatibles/química , Cerámica/química , Dióxido de Silicio/química , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/fisiología , Calcio/metabolismo , Humanos , Ensayo de Materiales , Tamaño de la Partícula , Porosidad , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA