Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Healthc Mater ; : e2401099, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38814677

RESUMEN

Endovascular embolization is a promising therapeutic approach broadening its application area due to its minimal invasiveness and short operation time, wherein lesional blood vessels are occluded with liquid embolic agents under X-ray imaging guidance. Histoacryl and its composition with Lipiodol are one of the most widely used liquid embolic agents, however, Histoacryl has critical limitations such as lack of innate X-ray visibility and strong adhesion to microcatheter. In this study, three different iodinated cyanoacrylates are newly synthesized as alternatives to Histoacryl and employed to develop liquid embolic compositions. Among them, 4-iodobutyl 2-cyanoacrylate (IBCA) was most preferable with high iodine content (730 mgI/mL) and fast polymerization. The IBCA-based embolic compositions containing ethyl oleate and acetic acid showed moderate viscosity and reduced catheter adhesiveness (∼ 0.80 N), and their polymerization time was freely controllable from 2 to 15 s. In the embolization test with rabbit models, the renal artery was successfully occluded by IBCA-based embolic compositions without vascular recanalization or non-target embolization for 4 w. Their embolic effect was further evaluated using swine models, demonstrating the practical applicability in the clinic. In conclusion, IBCA and its compositions are determined to have great potential as novel liquid embolic agents. This article is protected by copyright. All rights reserved.

2.
Biomolecules ; 13(12)2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38136656

RESUMEN

The advancement of stem cell therapy has offered transformative therapeutic outcomes for a wide array of diseases over the past decades. Consequently, stem cell tracking has become significant in revealing the mechanisms of action and ensuring safe and effective treatments. Fluorescence stands out as a promising choice for stem cell tracking due to its myriad advantages, including high resolution, real-time monitoring, and multi-fluorescence detection. Furthermore, combining fluorescence with other tracking modalities-such as bioluminescence imaging (BLI), positron emission tomography (PET), photoacoustic (PA), computed tomography (CT), and magnetic resonance (MR)-can address the limitations of single fluorescence detection. This review initially introduces stem cell tracking using fluorescence imaging, detailing various labeling strategies such as green fluorescence protein (GFP) tagging, fluorescence dye labeling, and nanoparticle uptake. Subsequently, we present several combinations of strategies for efficient and precise detection.


Asunto(s)
Células Madre Mesenquimatosas , Tomografía de Emisión de Positrones , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X , Imagen Multimodal
3.
Biomater Res ; 27(1): 102, 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37845762

RESUMEN

BACKGROUND: Nano-sized drug delivery system has been widely studied as a potential technique to promote tumor-specific delivery of anticancer drugs due to its passive targeting property, but resulting in very restricted improvements in its systemic administration so far. There is a requirement for a different approach that dramatically increases the targeting efficiency of therapeutic agents at targeted tumor tissues. METHODS: To improve the tumor-specific accumulation of anticancer drugs and minimize their undesirable toxicity to normal tissues, a tumor-implantable micro-syringe chip (MSC) with a drug reservoir is fabricated. As a clinically established delivery system, six liposome nanoparticles (LNPs) with different compositions and surface chemistry are prepared and their physicochemical properties and cellular uptake are examined in vitro. Subsequently, MSC-guided intratumoral administration is studied to identify the most appropriate for the higher tumor targeting efficacy with a uniform intratumoral distribution. For efficient cancer treatment, pro-apoptotic anticancer prodrugs (SMAC-P-FRRG-DOX) are encapsulated to the optimal LNPs (SMAC-P-FRRG-DOX encapsulating LNPs; ApoLNPs), then the ApoLNPs are loaded into the 1 µL-volume drug reservoir of MSC to be delivered intratumorally for 9 h. The tumor accumulation and therapeutic effect of ApoLNPs administered via MSC guidance are evaluated and compared to those of intravenous and intratumoral administration of ApoLNP in 4T1 tumor-bearing mice. RESULTS: MSC is precisely fabricated to have a 0.5 × 4.5 mm needle and 1 µL-volume drug reservoir to achieve the uniform intratumoral distribution of LNPs in targeted tumor tissues. Six liposome nanoparticles with different compositions of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (PC), 1,2-dioleoyl-sn-glycero-3-phospho-L-serine (PS), 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)2000] (PEG2000-DSPE) are prepared with average sizes of 100-120 nm and loaded into the 1 µL-volume drug reservoir in MSC. Importantly negatively charged 10 mol% of PS-containing LNPs are very slowly infused into the tumor tissue through the micro-syringe of the MSC over 6 h. The intratumoral targeting efficiency of MSC guidance is 93.5%, effectively assisting the homogeneous diffusion of LNPs throughout the tumor tissue at 3.8- and 2.7-fold higher concentrations compared to the intravenous and intratumoral administrations of LNPs, respectively. Among the six LNP candidates 10 mol% of PS-containing LNPs are finally selected for preparing pro-apoptotic SMAC-P-FRRG-DOX anticancer prodrug-encapsulated LNPs (ApoLNPs) due to their moderate endocytosis rate high tumor accumulation and homogenous intratumoral distribution. The ApoLNPs show a high therapeutic effect specifically to cathepsin B-overexpressing cancer cells with 6.6 µM of IC50 value while its IC50 against normal cells is 230.7 µM. The MSC-guided administration of ApoLNPs efficiently inhibits tumor growth wherein the size of the tumor is 4.7- and 2.2-fold smaller than those treated with saline and intratumoral ApoLNP without MSC, respectively. Moreover, the ApoLNPs remarkably reduce the inhibitor of apoptosis proteins (IAPs) level in tumor tissues confirming their efficacy even in cancers with high drug resistance. CONCLUSION: The MSC-guided administration of LNPs greatly enhances the therapeutic efficiency of anticancer drugs via the slow diffusion mechanism through micro-syringe to tumor tissues for 6 h, whereas they bypass most hurdles of systemic delivery including hepatic metabolism, rapid renal clearance, and interaction with blood components or other normal tissues, resulting in the minimum toxicity to normal tissues. The negatively charged ApoLNPs with cancer cell-specific pro-apoptotic prodrug (SMAC-P-FRRG-DOX) show the highest tumor-targeting efficacy when they are treated with the MSC guidance, compared to their intravenous or intratumoral administration in 4T1 tumor-bearing mice. The MSC-guided administration of anticancer drug-encapsulated LNPs is expected to be a potent platform system that facilitates overcoming the limitations of systemic drug administration with low delivery efficiency and serious side effects.

4.
Nanomaterials (Basel) ; 13(15)2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37570543

RESUMEN

Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.

5.
Pharmaceutics ; 15(2)2023 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-36839734

RESUMEN

Proteolysis-targeting chimeras (PROTACs) are rapidly emerging as a potential therapeutic strategy for cancer therapy by inducing the degradation of tumor-overexpressing oncogenic proteins. They can specifically catalyze the degradation of target oncogenic proteins by recruiting E3 ligases and utilizing the ubiquitin-proteasome pathway. Since their mode of action is universal, irreversible, recyclable, long-lasting, and applicable to 'undruggable' proteins, PROTACs are gradually replacing the role of conventional small molecular inhibitors. Moreover, their application areas are being expanded to cancer immunotherapy as various types of oncogenic proteins that are involved in immunosuppressive tumor microenvironments. However, poor water solubility and low cell permeability considerably restrict the pharmacokinetic (PK) property, which necessitates the use of appropriate delivery systems for cancer immunotherapy. In this review, the general characteristics, developmental status, and PK of PROTACs are first briefly covered. Next, recent studies on the application of various types of passive or active targeting delivery systems for PROTACs are introduced, and their effects on the PK and tumor-targeting ability of PROTACs are described. Finally, recent drug delivery systems of PROTACs for cancer immunotherapy are summarized. The adoption of an adequate delivery system for PROTAC is expected to accelerate the clinical translation of PROTACs, as well as improve its efficacy for cancer therapy.

6.
Biomaterials ; 295: 122038, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36787659

RESUMEN

Proteolysis-targeting chimeras (PROTACs) have recently been of great interest in cancer therapy. However, the bioavailability of PROTACs is considerably restricted due to their high hydrophobicity, poor cell permeability, and thereby low tumor targeting ability. Herein, esterase-cleavable maleimide linker (ECMal)-conjugated bromodomain 4 (BRD4)-degrading PROTAC (ECMal-PROTAC) is newly synthesized to exploit plasma albumin as an 'innate drug carrier' that can be accumulated in targeted tumor tissues. The BRD4-degrading ECMal-PROTAC is spontaneously bound to albumins via the thiol-maleimide click chemistry and its esterase-specific cleavage of ECMal-PROTAC is characterized in physiological conditions. The albumin-bound ECMal-PROTACs (Alb-ECMal-PROTACs) have an average size of 6.99 ± 1.38 nm, which is similar to that of free albumins without denaturation or aggregation. When Alb-ECMal-PROTACs are treated to 4T1 tumor cells, they are actively endocytosed and reach their highest intracellular level within 12 h. Furthermore, the maleimide linkers of Alb-ECMal-PROTACs are cleaved by the esterase to release free BRD-4 degrading PROTACs and the cell-internalized PROTACs successfully catalyze the selective degradation of BRD4 proteins, resulting in BRD4 deficiency-related apoptosis. When ECMal-PROTACs are intravenously injected into tumor-bearing mice, they exhibit a 16.3-fold higher tumor accumulation than free BRD4-PROTAC, due to the shuttling effect of albumin for tumor targeting. Finally, ECMal-PROTACs show 5.3-fold enhanced antitumor efficacy compared to free BRD4-PROTAC, without provoking any severe systemic toxicity. The expression of Bcl-2 and c-Myc, the downstream oncogenic proteins of BRD4, are also effectively suppressed. In summary, the in situ albumin binding of ECMal-PROTAC is proven as a promising strategy that effectively modulates its pharmacokinetics and therapeutic performance with high applicability to other types of PROTACs.


Asunto(s)
Neoplasias , Proteínas Nucleares , Animales , Ratones , Albúminas/metabolismo , Esterasas/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Quimera Dirigida a la Proteólisis
7.
Biomaterials ; 290: 121841, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36206664

RESUMEN

Immune checkpoint blockade (ICB) has shown remarkable therapeutic efficacy in a variety of cancers. However, patients exhibit unexpectedly low response rates to ICB therapy owing to the unwanted recycling and cellular abundance of PD-L1. Herein, rational design of PD-L1 multivalent binding liposome is investigated through PEGylated liposomes incorporating different ratios of PD-L1 binding peptide. Liposomes incorporating 10 mol% PD-L1 binding peptides (10-PD-L1-Lipo) promote the multivalent binding with PD-L1 on tumor cell surface, which is endocytosed for its trafficking toward the lysosomes instead of the recycling endosomes. Thereby, 10-PD-L1-Lipo leads to a significant PD-L1 degradation that prevents its recycling and cellular abundance compared to anti-PD-L1 antibody, disrupting immune escape mechanism of tumor cells and enhancing T cell-mediated antitumor immunity. Moreover, a clinically applicable doxorubicin (DOX) liposomal formulation is established via drug encapsulation into 10-PD-L1-Lipo. The resulting DOX-PD-L1-Lipo primes tumors via immunogenic chemotherapy by preferential DOX accumulation by the EPR effect and overcomes PD-L1 abundance induced following chemotherapy through multivalent binding-mediated PD-L1 degradation. As a result, the synergistic immunogenic chemotherapy and multivalent binding-mediated PD-L1 degradation by DOX-PD-L1-Lipo show significantly enhanced antitumor efficacy and immune responses in colon tumor models. Collectively, this study suggests the rationally designed PEGylated liposomes to promote PD-L1 multivalent binding providing a new route for safe and more effective ICB therapy.


Asunto(s)
Liposomas , Lisosomas , Humanos , Línea Celular Tumoral , Polietilenglicoles , Linfocitos T , Inmunoterapia/métodos
8.
Biomaterials ; 289: 121806, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36156411

RESUMEN

A carrier-free prodrug nanoparticle has emerged as a potential approach to cancer therapy. It plays a vital role in enhancing the tumor targeting and therapeutic efficacy of the anticancer agent at sites of intention wherein the prodrug nanoparticle is potentially activated. Herein, five derivatives of cathepsin B-cleavable prodrugs are synthesized via chemically conjugating different cathepsin B-cleavable peptides (Phe-Arg-Arg-Gly, Phe-Arg-Arg-Leu, Phe-Arg-Arg-Leu-Gly, Phe-Leu-Arg-Arg-Gly) to doxorubicin (DOX). The peptide-DOX prodrugs can spontaneously assemble into nanoparticles via their intermolecular hydrophobic and π-π stacking interactions. The resulting cathepsin B-cleavable prodrugs nanoparticles formed different nanoparticle structures according to the amphiphilicity and flexibility of different peptides and their particle stability and cellular uptake mechanism are carefully evaluated in vitro. Among five prodrug nanoparticles, the Phe-Arg-Arg-Leu-DOX (FRRL-DOX) nanoparticle was formed to a size of 167.5 ± 12.4 nm and stably maintains its nanoparticle structure in saline media for 3 days. The FRRL-DOX nanoparticle is well taken up by tumoral nuclei and effectively induces cancer cell death with minimal toxicity to normal cells. In addition, the FRRL-DOX nanoparticle shows 2.3-16.3-fold greater tumor-specific accumulation in vivo than other prodrug nanoparticles and free DOX. The therapeutic effect of FRRL-DOX is finally examined, demonstrating 2.1-fold better anticancer efficacy compared to that of free DOX. Notably, the FRRL-DOX nanoparticle does not exert serious toxicity in its repeated intravenous administration at a high dose of up to 10 mg/kg (equiv. to DOX). In conclusion, the peptide sequence for cathepsin B-cleavable prodrug nanoparticle is determined to be successfully optimized in a way of increasing its tumor selectivity and lowering toxicity to normal tissues.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Profármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Catepsina B/metabolismo , Catepsina B/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Profármacos/química
9.
Expert Opin Drug Deliv ; 19(6): 641-652, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35603410

RESUMEN

INTRODUCTION: Immune checkpoint blockade (ICB) therapy is now FDA-approved for the treatment of various tumor types. By removing inhibitory signals for T-cell activation and disrupting the immune escape mechanism of tumor cells, ICB therapy has shown considerable efficacy with complete tumor regression in patients. However, patients respond poorly to this therapy and show limited response rates owing to the immunosuppressive tumor microenvironment (ITM) in cold tumors. AREAS COVERED: In this review, recent advances and progress in the use of nano-sized drug delivery system (Nano-DDS) to potentiate the ICB therapy by reversing cold tumors with an ITM into immunogenic hot tumors are discussed. The types of immunogenic cell death (ICD) inducers that initiate or enhance antitumor immune responses are classified, and their extensive combination with immune modulators using Nano-DDS is highlighted. EXPERT OPINION: Nano-DDS can be efficiently combined with ICD inducers and immune modulators and trigger a potent antitumor immune response based on a comprehensive approach to the cancer-immunity cycle.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Sistema de Administración de Fármacos con Nanopartículas , Neoplasias/terapia , Microambiente Tumoral
10.
Pharmaceutics ; 14(4)2022 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-35456562

RESUMEN

Albumin has shown remarkable promise as a natural drug carrier by improving pharmacokinetic (PK) profiles of anticancer drugs for tumor-targeted delivery. The exogenous or endogenous albumin enhances the circulatory half-lives of anticancer drugs and passively target the tumors by the enhanced permeability and retention (EPR) effect. Thus, the albumin-based drug delivery leads to a potent antitumor efficacy in various preclinical models, and several candidates have been evaluated clinically. The most successful example is Abraxane, an exogenous human serum albumin (HSA)-bound paclitaxel formulation approved by the FDA and used to treat locally advanced or metastatic tumors. However, additional clinical translation of exogenous albumin formulations has not been approved to date because of their unexpectedly low delivery efficiency, which can increase the risk of systemic toxicity. To overcome these limitations, several prodrugs binding endogenous albumin covalently have been investigated owing to distinct advantages for a safe and more effective drug delivery. In this review, we give account of the different albumin-based drug delivery systems, from laboratory investigations to clinical applications, and their potential challenges, and the outlook for clinical translation is discussed. In addition, recent advances and progress of albumin-binding drugs to move more closely to the clinical settings are outlined.

11.
J Biomater Sci Polym Ed ; 33(1): 57-76, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34503403

RESUMEN

Co-delivery of microbubbles (MBs) with anticancer drugs is a promising theranostic approach that can enhance both the ultrasound contrast and local extravasation of drugs with the sonoporation effect. The simultaneous administration of MBs and hydrophobic drugs, however, is still challenging due to the limitations in drug loading or undesirable stabilization of MBs. In this research, MB-self-aggregate complexes (MB-SAs) were newly fabricated for the encapsulation of hydrophobic drugs, and their theranostic properties are investigated in vitro and in vivo. Glycol chitosan self-aggregates (GC-SAs) loaded with hydrophobic drugs or dyes were chemically conjugated on the surface MBs. Their conjugation ratio was determined to be 73.9%, and GC-SAs on MBs did not affect the stability of MBs. GC-SA attached MBs (GC@MBs) were successfully visualized with low-intensity insonation and showed enhanced cellular uptake via the sonoporation effect. In vivo biodistribution of GC@MBs was examined with tumor-bearing mice, confirming that their accumulation at the tumor site increased by 1.85 times after ultrasound irradiation. The anticancer drug-loaded GC@MBs also exhibited 10% higher cytotoxicity under ultrasound flash. In conclusion, it was expected that GC@MBs could be used both as an ultrasound contrast agent and a drug carrier even with conventional ultrasonic devices.


Asunto(s)
Sistemas de Liberación de Medicamentos , Microburbujas , Animales , Línea Celular Tumoral , Portadores de Fármacos , Ratones , Distribución Tisular
12.
J Biomater Sci Polym Ed ; 33(4): 409-425, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34613885

RESUMEN

Transarterial chemoembolization (TACE) is a therapeutic approach to address hepatocellular carcinoma by obstructing the blood supply to the tumor using embolic agents and improving the local delivery of anticancer agents. Size-calibrated polymeric microspheres (MSs) termed drug-eluting beads (DEBs) are the most prevalent solid embolic materials; however, their limitations include insufficient X-ray visibility or biodegradability. In this study, size-controlled polymeric MSs with inherent radiopacity and biodegradability were created, and their embolic effect was assessed. Poly(lactide-co-glycolide) MSs (PLGA MSs) incorporating a hydrophobic X-ray contrast agent and an anticancer drug were produced by the w/o/w emulsion process. Their sizes were exactly calibrated to 71.40 ± 32.18 and 142.66 ± 59.92 µm in diameter, respectively, which were confirmed to have sizes similar to the clinically available DEBs. The iodine content of PLGA MSs was calculated as 144 mgI/g, and the loading quantity of the drug was 1.33%. Manufactured PLGA MSs were gradually degraded for 10 weeks and consistently released the anticancer drug. Following the PLGA MSs injection into the renal artery of New Zealand white rabbit test subjects, their deliverability to the targeted vessel through the microcatheter was confirmed. Injected PLGA MSs were clearly imaged through the real-time X-ray device without blending any contrast agents. The embolic effect of the PLGA MSs was ultimately established by the atrophy of an embolized kidney after 8 weeks. Consequently, the designed PLGA MS is anticipated to be an encouraging prospect to address hepatocellular carcinoma.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Quimioembolización Terapéutica , Neoplasias Hepáticas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/terapia , Medios de Contraste , Humanos , Ácido Láctico/química , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/terapia , Microesferas , Poliglactina 910 , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Conejos
13.
Acta Biomater ; 131: 286-301, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34246803

RESUMEN

Injectable hydrogels have been studied as drug delivery systems because of their minimal invasiveness and sustained drug release properties. Pluronic F127, consisting of poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) triblock copolymers, exhibits thermo-responsive properties and hence is injectable due to its rapid sol-gel transition. Unmodified Pluronic F127-based hydrogels, however, have limited long-term stability and controllable release of drugs entrapped within them. In this study, host-guest interactions between adamantane-conjugated Pluronic F127 (F127-Ad) and polymerized ß-cyclodextrin (CDP) were employed to develop a hydrogel-based protein delivery system. Single or multiple adamantane units were successfully introduced at the termini of Pluronic F127 with a 100% conversion yield, and the synthesized F127-Ad polymer produced a physically crosslinked micelle-packing structure when mixed with CDP. As the number of adamantanes at the terminal ends of Pluronic F127 increased, the critical gelation concentration of F127-Ad/CDP hydrogel decreased from 15 to 6% (w/v). The F127/CDP hydrogel was able to maintain its structure even with lower polymer content, and its injectability improved with a reduction of the hydrogel viscosity. The long-term stability of F127/CDP hydrogels was evaluated in vitro and in vivo, and it was demonstrated that the subcutaneously injected hydrogel did not disintegrate for up to 30 d. Throughout the drug release test using gelatin and insulin as model drugs, it was demonstrated that their release rates could be regulated via complexation between the protein drugs and the ß-cyclodextrin molecules inside the hydrogel. In conclusion, the F127-Ad/CDP hydrogel is expected to be a versatile protein delivery system with controllable durability and drug release characteristics. STATEMENT OF SIGNIFICANCE: Pluronic F127 is one of the widely studied polymeric materials for thermo-sensitive injectable hydrogels due to its high biocompatibility and rapid sol-gel transition. Since the Pluronic F127-based hydrogel has some limitations in its long-term stability and mechanical property, it is inevitable to modify its structure for the application to drug delivery. In this study, mono- or multi- adamantane-conjugated Pluronic F127s were synthesized and mixed with ß-cyclodextrin polymers to form hydrogels with host-guest interaction-mediated micelle-packing structures. The host-guest interaction introduced into the hydrogel system endowed it a sustained protein drug release behavior as well as high durability in vitro and in vivo. By increasing the number of adamantane molecules at the end of the Pluronic F127, both the stability and injectability of the hydrogel could be also modulated.


Asunto(s)
Hidrogeles , Poloxámero , Liberación de Fármacos , Micelas , Proteínas , Temperatura
14.
J Mater Chem B ; 9(11): 2631-2640, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33683280

RESUMEN

Islet cell transplantation has been an effective method for the treatment of type 1 diabetes. The transplanted islets release insulin in response to changes in blood glucose levels. The clinical application of islet transplantation, however, has been hindered because of some critical problems including immune responses to grafted islets and side effects caused by overdosed immunosuppressive drugs. Herein, surface modification technology using poly(ethylene glycol) (PEG)-dendron was proposed to safeguard islets from the host immune system. PEG-dendron was synthesized by a divergent polymerization method and utilized to cover the islet antigen surface. Successful conjugation of PEG-dendron on the islet surface was achieved without affecting islet morphology, viability, and functionality at a concentration of 1.00%. Surface modification using PEG-dendron effectively prevented protein absorption and immune activation. Foremost, it improved the survival rate of islet grafts in vivo when combined with a low dose of immunosuppressive drugs. In conclusion, PEG-dendron is a potential candidate for the surface modification of pancreatic islets to mitigate immune responses after transplantation.


Asunto(s)
Dendrímeros/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Inmunosupresores/farmacología , Islotes Pancreáticos/efectos de los fármacos , Polietilenglicoles/farmacología , Animales , Dendrímeros/química , Diabetes Mellitus Experimental/inmunología , Inmunosupresores/química , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Ratas , Ratas Sprague-Dawley
15.
Adv Drug Deliv Rev ; 76: 60-78, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25064554

RESUMEN

Molecular imaging non-invasively visualizes and characterizes the biologic functions and mechanisms in living organisms at a molecular level. In recent years, advances in imaging instruments, imaging probes, assay methods, and quantification techniques have enabled more refined and reliable images for more accurate diagnoses. Multimodal imaging combines two or more imaging modalities into one system to produce details in clinical diagnostic imaging that are more precise than conventional imaging. Multimodal imaging offers complementary advantages: high spatial resolution, soft tissue contrast, and biological information on the molecular level with high sensitivity. However, combining all modalities into a single imaging probe involves problems yet to be solved due to the requirement of high dose contrast agents for a component of imaging modality with low sensitivity. The introduction of targeting moieties into the probes enhances the specific binding of targeted multimodal imaging modalities and selective accumulation of the imaging agents at a disease site to provide more accurate diagnoses. An extensive list of prior reports on the targeted multimodal imaging probes categorized by each modality is presented and discussed. In addition to accurate diagnosis, targeted multimodal imaging agents carrying therapeutic medications make it possible to visualize the theranostic effect and the progress of disease. This will facilitate the development of an imaging-guided therapy, which will widen the application of the targeted multimodal imaging field to experiments in vivo.


Asunto(s)
Imagen Multimodal/métodos , Animales , Humanos , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X
16.
ACS Macro Lett ; 3(11): 1112-1116, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-35610806

RESUMEN

Supramolecular assembly of end-functionalized polymers, forming block copolymer-like supramolecules based on ionic interaction, has been utilized as a simple and facile method for generating functionalized nanoporous thin film. Here, the binary blend film of aminated poly(ethylene oxide) dendrimer (APEO-G) and sulfonated polystyrene (SPS) at a stoichiometric composition after benzene/water solvent vapor annealing exhibits spherical domains in multilayers over a large area. By controlling the number of end-functional arms of dendrimer via divergent ring-opening polymerization of ethylene oxide as well as the molecular weights of SPS, the domain sizes can be controlled ranging from mainly 34 to 54 nm, even to 131 nm. Our supramolecular-assembly system provides an alternative approach to fabricating a functional nanotemplate by easily etching domains with selective solvent treatment and leaving functional groups at the pore surfaces.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...