Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 18(1): 199-209, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38109681

RESUMEN

Tumor-derived extracellular vesicles (TDEs) have potential for therapeutic cancer vaccine applications since they innately possess tumor-associated antigens, mediate antigen presentation, and can incorporate immune adjuvants for enhanced vaccine efficacy. However, the original TDEs also contain immune-suppressive proteins. To address this, we proposed a simple yet powerful preconditioning method to improve the overall immunogenicity of the TDEs. This approach involved inducing endoplasmic reticulum (ER) stress on parental tumor cells via N-glycosylation inhibition with tunicamycin. The generated immunogenic TDEs (iTDEs) contained down-regulated immunosuppressive proteins and up-regulated immune adjuvants, effectively activating dendritic cells (DCs) in vitro. Furthermore, in vivo evidence from a tumor-bearing mouse model showed that iTDEs activated DCs, enabling cytotoxic T lymphocytes (CTLs) to target tumors, and eventually established a systemic antitumor immune response. Additionally, iTDEs significantly delayed tumor recurrence in a postsurgery model compared with control groups. These findings highlight the immense potential of our strategy for utilizing TDEs to develop effective cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Vesículas Extracelulares , Neoplasias , Ratones , Animales , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/terapia , Linfocitos T Citotóxicos , Adyuvantes Inmunológicos , Retículo Endoplásmico , Células Dendríticas
2.
Small ; 18(36): e2106569, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35322545

RESUMEN

The unique biological characteristics and promising clinical potential of extracellular vesicles (EVs) have galvanized EV applications for regenerative medicine. Recognized as important mediators of intercellular communication, naturally secreted EVs have the potential, as innate biotherapeutics, to promote tissue regeneration. Although EVs have emerged as novel therapeutic agents, challenges related to the clinical transition have led to further functionalization. In recent years, various engineering approaches such as preconditioning, drug loading, and surface modification have been developed to potentiate the therapeutic outcomes of EVs. Also, limitations of natural EVs have been addressed by the development of artificial EVs that offer advantages in terms of production yield and isolation methodologies. In this review, an updated overview of current techniques is provided for the functionalization of natural EVs and recent advances in artificial EVs, particularly in the scope of regenerative medicine.


Asunto(s)
Vesículas Extracelulares , Medicina Regenerativa , Transporte Biológico , Vesículas Extracelulares/metabolismo , Medicina Regenerativa/métodos
3.
Int J Biol Macromol ; 192: 1231-1239, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34626726

RESUMEN

Anti-death receptor 5 (DR5) antibody is a potential therapeutic agent for liver fibrosis because it exhibits anti-fibrotic effects by inducing the apoptosis of activated hepatic stellate cells (HSCs), which are responsible for hepatic fibrogenesis. However, the clinical applications of anti-DR5 antibodies have been limited by their low agonistic activity against DR5. In this study, an anti-DR5 antibody-curcumin conjugate (DCC) was prepared to investigate its effect on the clearance of activated HSCs. The DCC was synthesized through a coupling reaction between a maleimide-functionalized curcumin derivative and a thiolated anti-DR5 antibody. No significant differences were observed in the uptake behaviors of activated HSCs between the bare anti-DR5 antibodies and DCC. Owing to the antioxidant and anti-inflammatory effects of curcumin, DCC-treated HSCs produced much lower levels of reactive oxygen species and inducible nitric oxide synthase than the bare anti-DR5 antibody-treated HSCs. Additionally, the anti-fibrotic effects of DCC on activated HSCs were more prominent than those of the bare anti-DR5 antibodies, as demonstrated by the immunocytochemical analysis of α-smooth muscle actin. DCC preferentially accumulated in the liver after its systemic administration to mice with liver fibrosis. Thus, DCC may serve as a potential therapeutic agent for treating liver fibrosis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Curcumina/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Inmunoconjugados/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Química Sintética , Células Estrelladas Hepáticas/metabolismo , Inmunoconjugados/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular
4.
Sci Adv ; 7(23)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34078596

RESUMEN

Despite the remarkable advances in therapeutics for rheumatoid arthritis (RA), a large number of patients still lack effective countermeasures. Recently, the reprogramming of macrophages to an immunoregulatory phenotype has emerged as a promising therapeutic strategy for RA. Here, we report metabolically engineered exosomes that have been surface-modified for the targeted reprogramming of macrophages. Qualified exosomes were readily harvested from metabolically engineered stem cells by tangential flow filtration at a high yield while maintaining their innate immunomodulatory components. When systemically administered into mice with collagen-induced arthritis, these exosomes effectively accumulated in the inflamed joints, inducing a cascade of anti-inflammatory events via macrophage phenotype regulation. The level of therapeutic efficacy obtained with bare exosomes was achievable with the engineered exosomes of 10 times less dose. On the basis of the boosted nature to reprogram the synovial microenvironment, the engineered exosomes display considerable potential to be developed as a next-generation drug for RA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Exosomas , Animales , Artritis Experimental/terapia , Artritis Reumatoide/tratamiento farmacológico , Humanos , Macrófagos , Ratones , Células Madre
5.
J Control Release ; 330: 15-30, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33278480

RESUMEN

The biological significance of extracellular vesicles (EVs) as intercellular communication mediators has been increasingly revealed in a wide range of normal physiological processes and disease pathogenesis. In particular, regenerative and immunomodulatory EVs hold potential as innate biotherapeutics, whereas pathological EVs are considered therapeutic targets for inhibiting their bioactivity. Given their ability to transport functional cargos originating from the source cells to target cells, EVs can also be used as a therapeutic means to deliver drug molecules. This review aims to provide an updated overview of the key engineering approaches for better exploiting EVs in disease intervention. The emphasis is lying on the preconditioning methods for therapeutic EVs, drug loading and targeting technologies for carrier EVs, and activity control strategies for pathological EVs.


Asunto(s)
Vesículas Extracelulares , Comunicación Celular , Sistemas de Liberación de Medicamentos
6.
Sci Adv ; 6(21): eaaz8400, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32637587

RESUMEN

Chemiluminescence (CL) has recently gained attention for CL resonance energy transfer (CRET)-mediated photodynamic therapy of cancer. However, the short duration of the CL signal and low quantum yield of the photosensitizer have limited its translational applications. Here, we report CRET-based nanoparticles (CRET-NPs) to achieve quantum yield-enhanced cancer phototheranostics by reinterpreting the hidden nature of CRET. Owing to reactive oxygen species (ROS)-responsive CO2 generation, CRET-NPs were capable of generating a strong and long-lasting photoacoustic signal in the tumor tissue via thermal expansion-induced vaporization. In addition, the CRET phenomenon of the NPs enhanced ROS quantum yield of photosensitizer through both electron transfer for an oxygen-independent type I photochemical reaction and self-illumination for an oxygen-dependent type II photochemical reaction. Consequently, owing to their high ROS quantum yield, CRET-NPs effectively inhibited tumor growth with complete tumor growth inhibition in 60% of cases, even with a single treatment.

7.
Acc Chem Res ; 52(7): 1771-1782, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31241894

RESUMEN

Growth in the knowledge of cancer biology has led to the emergence and evolution of cancer nanomedicines by providing the rationale for leveraging nanotechnology to develop better treatment options. The discovery of nanometer-sized intercellular openings in the defective angiogenic tumor vasculature contributed to the development of an idea for the well-known cancer passive targeting regime, enhanced permeability and retention (EPR) effect, of the nanomedicines. Recently, reactive oxygen species (ROS) have been highlighted as one of the key players that underlie the acquisition of the various hallmarks of cancer. As ROS are associated with all stages of cancer, their applications in cancer treatment based on the following concentration-dependent implications have attracted much attention: (1) low to moderate levels of ROS as key signaling molecules, (2) elevated levels of ROS in cancer cells as one of the unique characteristics of cancer, and (3) excessive levels of ROS as cytotoxic agents. Considering ROS from a different point of view, various cancer nanomedicines have been designed to achieve spatiotemporal control of therapeutic action, the main research focus in this area. This Account includes our efforts and preclinical achievements in development of nanomedicines for a range of ROS-mediated cancer therapies. It begins with general background regarding cancer nanomedicines, the significance of ROS in cancer, and a brief overview of ROS-mediated approaches for cancer therapy. Then, this Account highlights the two key roles of ROS that define therapeutic purposes of cancer nanomedicines: (1) ROS as drug delivery enhancers and (2) ROS as cell death inducers. The former inspired us to develop nitric oxide-generating nanoparticles for improved EPR effect, endogenous ROS-responsive polymeric micelles for enhanced intracellular drug delivery, and exogenous ROS-activated micelles for subcellular localization via photochemical internalization. While refining conventional chemotherapy, recent researches also have focused on the latter, the cytotoxic ROS, to advance alternative treatment modalities such as oxidation therapy, photodynamic therapy (PDT), and sonodynamic therapy (SDT). In particular, we have been motivated to develop polymeric nanoreactors containing enzymes to produce H2O2 for oxidation therapy, photosensitizer-loaded gold-nanoclustered polymeric nanoassemblies for photothermally activated PDT overcoming the oxygen dependency of PDT, and hydrophilized TiO2 nanoparticles and Au-TiO2 nanocomposites as novel sonosensitizers for improved SDT efficiency. The integration of nanomedicine and ROS-mediated therapy has emerged as the new paradigm in the treatment of cancer, based on promising proof-of-concept demonstrations in preclinical studies. Further efforts to ensure clinical translation along with more sophisticated cancer nanomedicines to address relevant challenges are expected to be made in the coming years.


Asunto(s)
Antineoplásicos/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular Tumoral , Portadores de Fármacos/química , Glucosa Oxidasa/química , Humanos , Ratones , Nanomedicina/métodos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Terapia por Ultrasonido/métodos , Verteporfina/uso terapéutico
8.
Acta Biomater ; 79: 294-305, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30134209

RESUMEN

Since delivering drugs to an entire tumoral region leads to high therapeutic efficacy and good prognosis, achieving deep tumoral penetration of drugs is a major issue in cancer treatment. In this regard, conventional nanomedicines (>50 nm) have shown limitations in cancer therapy, primarily attributed to the heterogeneous distribution of drugs because of the physiological barrier of the tumor interstitial space. To address this issue, we prepared transformable hybrid nanoparticles (TNPs) consisting of a pH-responsive nanocarrier (PEG-PBAE) and doxorubicin (DOX)-conjugated ultrasmall (<3 nm) gold nanoparticles (nanosatellites). It has been shown that PEG-PBAE can serve as a reservoir for nanosatellites and release them in mildly acidic conditions (pH 6.5), mimicking the tumor microenvironment. When DOX-loaded TNPs (DOX-TNPs) were intravenously injected into tumor-bearing mice, they successfully accumulated and dissociated at the extracellular level of the tumor, leading to the disclosure of nanosatellites and free DOX. While the free DOX accumulated in tumor tissue near blood vessels, the deeply diffused nanosatellites were taken up by the tumor cell, followed by the release of DOX via cleavage of pH-responsive ester linkages in the nanosatellites at the intracellular level. Consequently, the DOX-TNPs effectively suppressed tumor growth through improved tumor penetration of DOX, suggesting their promising potential as a cancer nanomedicine. STATEMENT OF SIGNIFICANCE: Deep tumor penetration of anticancer drug is an important issue for high therapeutic efficacy. If the drugs cannot reach cancer cells in a sufficient concentration, their effectiveness will be limited. In this regard, conventional nanomedicine showed only modest therapeutic efficacy since they cannot deliver their payloads to the deep site of tumor tissue. This heterogeneous distribution of the drug is primarily attributed to the physiological barriers of the tumor microenvironment, including a dense extracellular matrix. To surmount this challenge, we developed tumor acidity-triggered transformable nanoparticles. By encapsulating doxorubicin-conjugated ultrasmall gold nanosatellites into the nanoparticles, the drug was not significantly bound to genetic materials, resulting in its minimal sequestration near the vasculature and deep tumor penetration. Our strategy could resolve not only the poor penetration issue of the drug but also its restricted tumor accumulation, suggesting the potential as an effective nanotherapeutics.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Neoplasias/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Doxorrubicina/farmacología , Liberación de Fármacos , Células HCT116 , Humanos , Nanopartículas del Metal/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Neoplasias/patología , Tamaño de la Partícula , Distribución Tisular/efectos de los fármacos
9.
J Control Release ; 279: 89-98, 2018 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29649530

RESUMEN

Self-assembled hyaluronic acid nanoparticles (HA-NPs) have been extensively investigated for biomedical and pharmaceutical applications owing to their biocompatibility and receptor-binding properties. Here, we report that an empty HA-NP itself not bearing any drug has therapeutic effects on adipose tissue inflammation and insulin resistance. HA-NPs inhibited not only the receptor-mediated internalization of low-molecular-weight (LMW) free HA but also LMW free HA-induced pro-inflammatory gene expression in mouse primary bone marrow-derived macrophages (BMDMs) isolated from wild-type mice, but not in CD44-null (CD44-/-) BMDMs. An in vivo biodistribution study showed the distribution of HA-NPs and their co-localization with CD44 in adipose tissues including epididymal white adipose tissues (eWATs), but these were rarely observed in the eWATs of CD44-/- mice. In addition, CD44 expression and HA-NP accumulation in the eWATs were increased in mice with diet-induced obesity (DIO) compared to lean mice. Interestingly, treatment with HA-NPs in DIO mice suppressed adipose tissue inflammation as indicated by reduced macrophage content, the production of proinflammatory cytokines and NLRP3 inflammasome activity in eWATs, leading to improved insulin sensitivity and normalized blood glucose levels. Collectively, these results suggest that an empty HA-NP itself can be a therapeutic agent for the treatment of type 2 diabetes.


Asunto(s)
Ácido Hialurónico/administración & dosificación , Inflamación/tratamiento farmacológico , Macrófagos/metabolismo , Nanopartículas/administración & dosificación , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Glucemia/efectos de los fármacos , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Receptores de Hialuranos/genética , Ácido Hialurónico/farmacocinética , Inflamación/patología , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Nanomedicina/métodos , Obesidad/metabolismo , Distribución Tisular
10.
Nano Lett ; 18(4): 2637-2644, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29521509

RESUMEN

Conventional cancer targeting with nanoparticles has been based on the assumed enhanced permeability and retention (EPR) effect. The data obtained in clinical trials to date, however, have rarely supported the presence of such an effect. To address this challenge, we formulated intracellular nitric oxide-generating nanoparticles (NO-NPs) for the tumor site-specific delivery of NO, a well-known vasodilator, with the intention of boosting EPR. These nanoparticles are self-assembled under aqueous conditions from amphiphilic copolymers of poly(ethylene glycol) and nitrated dextran, which possesses inherent NO release properties in the reductive environment of cancer cells. After systemic administration of the NO-NPs, we quantitatively assessed and visualized increased tumor blood flow as well as enhanced vascular permeability than could be achieved without NO. Additionally, we prepared doxorubicin (DOX)-encapsulated NO-NPs and demonstrated consequential improvement in therapeutic efficacy over the control groups with considerably improved DOX intratumoral accumulation. Overall, this proof of concept study implies a high potency of the NO-NPs as an EPR enhancer to achieve better clinical outcomes.

11.
Int J Biol Macromol ; 110: 406-415, 2018 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-29055700

RESUMEN

Trop2, a transmembrane glycoprotein, has emerged as a biomarker for targeted cancer therapy since it is overexpressed in 80% of triple negative breast cancer (TNBC) patients. For the site-specific delivery of the anticancer drug into TNBC, anti-Trop2 antibody-conjugated nanoparticles (ST-NPs) were prepared as the potential nanocarrier, composed of carboxymethyl dextran (CMD) derivatives with bioreducible disulfide bonds. Owing to its amphiphilicity, the CMD derivatives were self-assembled into nano-sized particles in an aqueous condition. Doxorubicin (DOX), chosen as a model anticancer drug, was effectively encapsulated into the nanoparticles. DOX-loaded ST-NPs (DOX-ST-NPs) rapidly released DOX in the presence of 10mM glutathione (GSH), whereas the DOX release is significantly retarded in the physiological condition (PBS, pH 7.4). Confocal microscopic images and flow cytometry analysis demonstrated that DOX-ST-NPs were selectively taken up by MDA-MB-231 as the representative Trop2-expressing TNBC cells. Consequently, DOX-ST-NPs exhibited higher toxicity to Trop2-positive MDA-MB-231 cancer cells, compared to DOX-loaded control nanoparticles without the disulfide bond or anti-Trop2 antibody. Overall, ST-NPs might be a promising carrier of DOX for targeted TNBC therapy.


Asunto(s)
Antineoplásicos Inmunológicos , Moléculas de Adhesión Celular/antagonistas & inhibidores , Dextranos , Doxorrubicina , Portadores de Fármacos , Nanopartículas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antígenos de Neoplasias , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Dextranos/química , Dextranos/farmacocinética , Dextranos/farmacología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Femenino , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Oxidación-Reducción , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
12.
J Control Release ; 267: 181-190, 2017 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-28844759

RESUMEN

The cell-free approach to foreignizing tumor cells with non-self antigens has received increasing attention as a method to induce cytotoxic T lymphocyte (CTL)-mediated immunological rejection of tumors, because the clinical translation of the conventional CTL-based cancer immunotherapies has been limited by a complicated manufacturing process and autotransplantation. In this study, we prepared matrix metalloproteinase 9 (MMP9)-responsive polymeric conjugates consisting of PEGylated hyaluronic acid (HA) as the targeting moiety and ovalbumin (OVA) as the model foreign antigen. The MMP9-cleavable linker was introduced between PEG and the HA backbone to facilitate the detachment of the PEG corona from the conjugate at the tumor site. From the in vitro cellular uptake study, it was revealed that the conjugate was effectively taken up by the CD44-expressing TC-1 cancer cells in the presence of MMP9 via receptor-mediated endocytosis. When the conjugate was systemically administered into the tumor-bearing mice with endogenous OVA-specific CTLs, the tumor growth was markedly inhibited, which was attributed to the significant antigen presentation on the tumor cells. Overall, the MMP9-responsive conjugates bearing foreign antigens might have the potential as an alternative to CTL-based cancer immunotherapeutics.


Asunto(s)
Antígenos/inmunología , Ácido Hialurónico/inmunología , Inmunoterapia , Metaloproteinasa 9 de la Matriz/inmunología , Neoplasias/terapia , Ovalbúmina/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos/química , Apoptosis , Línea Celular Tumoral , Femenino , Ácido Hialurónico/química , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Neoplasias/inmunología , Neoplasias/patología , Ovalbúmina/química , Polietilenglicoles/química
13.
Biomaterials ; 131: 15-26, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28371624

RESUMEN

With the aim of developing nanoparticles for targeted delivery of methotrexate (MTX) to inflamed joints in rheumatoid arthritis (RA), an amphiphilic polysaccharide was synthesized by conjugating 5ß-cholanic acid to a dextran sulfate (DS) backbone. Due to its amphiphilic nature, the DS derivative self-assembled into spherical nanoparticles (220 nm in diameter) in aqueous conditions. The MTX was effectively loaded into the DS nanoparticles (loading efficiency: 73.0%) by a simple dialysis method. Interestingly, the DS nanoparticles were selectively taken up by activated macrophages, which are responsible for inflammation and joint destruction, via scavenger receptor class A-mediated endocytosis. When systemically administrated into mice with experimental collagen-induced arthritis (CIA), the DS nanoparticles effectively accumulated in inflamed joints (12-fold more than wild type mice (WT)), implying their high targetability to RA tissues. Moreover, the MTX-loaded DS nanoparticles exhibited significantly improved therapeutic efficacy against CIA in mice compared to free MTX alone. Overall, the data presented here indicate that DS nanoparticles are potentially useful nanomedicines for RA imaging and therapy.


Asunto(s)
Antirreumáticos/administración & dosificación , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/tratamiento farmacológico , Sulfato de Dextran/análisis , Portadores de Fármacos/análisis , Metotrexato/administración & dosificación , Nanopartículas/análisis , Animales , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapéutico , Masculino , Metotrexato/farmacocinética , Metotrexato/uso terapéutico , Ratones , Imagen Óptica , Células RAW 264.7 , Nanomedicina Teranóstica
14.
J Mater Chem B ; 5(35): 7319-7327, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32264181

RESUMEN

Photodynamic therapy (PDT) has been extensively investigated to treat cancer since it induces cell death through the activation of photosensitizers by light. However, its success has been hampered by the insufficient selectivity of photosensitizers to tumor tissues. In an attempt to increase the therapeutic efficacy of PDT by targeting the photosensitizer specifically to the tumor site, we prepared chlorin e6 (Ce6)-loaded gold-stabilized carboxymethyl dextran nanoparticles (Ce6-GS-CNPs). Ce6-GS-CNPs possessed highly stable nanostructures and no significant change was observed in their particle size in the presence of serum for 6 days. When Ce6-GS-CNPs were intravenously injected into tumor-bearing mice, they exhibited prolonged circulation in the body and gradually accumulated in the tumor tissue. Under laser irradiation of the tumor site which could be recognized by the near-infrared fluorescence imaging system, Ce6-GS-CNPs effectively suppressed tumor growth. Overall, Ce6-GS-CNPs might have potential as nanomedicine for image-guided photodynamic cancer therapy.

15.
ACS Nano ; 10(12): 10858-10868, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-28024382

RESUMEN

Optically active nanomaterials have shown great promise as a nanomedicine platform for photothermal or photodynamic cancer therapies. Herein, we report a gold-nanoclustered hyaluronan nanoassembly (GNc-HyNA) for photothermally boosted photodynamic tumor ablation. Unlike other supramolecular gold constructs based on gold nanoparticle building blocks, this system utilizes the nanoassembly of amphiphilic hyaluronan conjugates as a drug carrier for a hydrophobic photodynamic therapy agent verteporfin, a polymeric reducing agent, and an organic nanoscaffold upon which gold can grow. Gold nanoclusters were selectively installed on the outer shell of the hyaluronan nanoassembly, forming a gold shell. Given the dual protection effect by the hyaluronan self-assembly as well as by the inorganic gold shell, verteporfin-encapsulated GNc-HyNA (Vp-GNc-HyNA) exhibited outstanding stability in the bloodstream. Interestingly, the fluorescence and photodynamic properties of Vp-GNc-HyNA were considerably quenched due to the gold nanoclusters covering the surface of the nanoassemblies; however, photothermal activation by 808 nm laser irradiation induced a significant increase in temperature, which empowered the PDT effect of Vp-GNc-HyNA. Furthermore, fluorescence and photodynamic effects were recovered far more rapidly in cancer cells due to certain intracellular enzymes, particularly hyaluronidases and glutathione. Vp-GNc-HyNA exerted a great potential to treat tumors both in vitro and in vivo. Tumors were completely ablated with a 100% survival rate and complete skin regeneration over the 50 days following Vp-GNc-HyNA treatment in an orthotopic breast tumor model. Our results suggest that photothermally boosted photodynamic therapy using Vp-GNc-HyNA can offer a potent therapeutic means to eradicate tumors.


Asunto(s)
Oro , Ácido Hialurónico , Nanopartículas del Metal , Fotoquimioterapia , Portadores de Fármacos , Humanos , Neoplasias/terapia
16.
Nano Lett ; 16(10): 6257-6264, 2016 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-27643533

RESUMEN

Although sonodynamic therapy (SDT) has emerged as a potential alternative to conventional photodynamic therapy, the low quantum yield of the sonosensitizer such as TiO2 nanoparticles (NPs) is still a major concern. Here, we have developed hydrophilized Au-TiO2 nanocomposites (HAu-TiO2 NCs) as sonosensitizers for improved SDT. The physicochemical properties of HAu-TiO2 NCs were thoroughly studied and compared with their counterparts without gold deposition. Upon exposure of HAu-TiO2 NCs to ultrasound, a large quantity of reactive oxygen species (ROS) were generated, leading to complete suppression of tumor growth after their systemic administration in vivo. Overall, it was evident that the composites of gold with TiO2 NPs significantly augmented the levels of ROS generation, implying their potential as SDT agents for cancer therapy.

17.
Biomaterials ; 103: 56-66, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27372421

RESUMEN

Stimuli-responsive micelles have emerged as the drug carrier for cancer therapy since they can exclusively release the drug via their structural changes in response to the specific stimuli of the target site. Herein, we developed the in situ diselenide-crosslinked micelles (DCMs), which are responsive to the abnormal ROS levels of tumoral region, as anticancer drug carriers. The DCMs were spontaneously derived from selenol-bearing triblock copolymers consisting of polyethylene glycol (PEG) and polypeptide derivatives. During micelle formation, doxorubicine (DOX) was effectively encapsulated in the hydrophobic core, and diselenide crosslinks were formed in the shell. The DCMs maintained their structural integrity, at least for 6 days in physiological conditions, even in the presence of destabilizing agents. However, ROS-rich conditions triggered rapid release of DOX from the DOX-encapsulating DCMs (DOX-DCMs) because the hydrophobic diselenide bond was cleaved into hydrophilic selenic acid derivatives. Interestingly, after their systemic administration into the tumor-bearing mice, DOX-DCMs delivered significantly more drug to tumors (1.69-fold and 3.73-fold higher amount compared with their non-crosslinked counterparts and free drug, respectively) and effectively suppressed tumor growth. Overall, our data indicate that DCMs have great potential as drug carriers for anticancer therapy.


Asunto(s)
Doxorrubicina/administración & dosificación , Doxorrubicina/química , Nanocápsulas/química , Neoplasias Experimentales/química , Neoplasias Experimentales/tratamiento farmacológico , Especies Reactivas de Oxígeno/química , Selenio/química , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Estabilidad de Medicamentos , Emulsiones/química , Humanos , Masculino , Ratones , Ratones Desnudos , Micelas , Terapia Molecular Dirigida/métodos , Nanocápsulas/administración & dosificación , Nanocápsulas/ultraestructura , Tamaño de la Partícula , Polietilenglicoles/química , Resultado del Tratamiento
18.
J Control Release ; 234: 68-78, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27164544

RESUMEN

In an attempt to develop the tumor-targeted nanocarrier which can surmount major challenges for in vivo application, we prepared tumor microenvironment-specific nanoparticles which can be sequentially activated at the extracellular and intracellular levels of tumor tissue by stepwise transformation. This polymeric nanoparticle has been prepared using an amphiphilic polyethyleneimine derivative with the pH-responsive charge-convertible moiety and the reduction-responsive crosslink. Once reaching the tumor tissue in vivo after systemic administration, the surface charge of this nanoparticle can be converted from negative to positive by recognizing the mildly acidic extracellular matrix of tumor, allowing for the enhanced cellular uptake. After the cellular uptake, the nanoparticle can selectively release the drug at the intracellular level since it has the chemically crosslinked core by the disulfide bond which is cleaved in intracellular reductive environment. The tumor microenvironment-specific nanoparticle shows the high tumor targetability and dramatically improves the antitumor efficacy of the drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Nanopartículas/química , Polietileneimina/química , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Propiedades de Superficie , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Biomaterials ; 27(1): 119-26, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16023198

RESUMEN

Self-assembled nanoparticles, formed by polymeric amphiphiles, have been demonstrated to accumulate in solid tumors by the enhanced permeability and retention effect, following intravenous administration. In this study, hydrophobically modified glycol chitosans capable of forming nano-sized self-aggregates were prepared by chemical conjugation of fluorescein isothiocyanate or doxorubicin to the backbone of glycol chitosan. Biodistribution of self-aggregates (300 nm in diameter) was evaluated using tissues obtained from tumor-bearing mice, to which self-aggregates were systemically administered via the tail vein. Irrespective of the dose, a negligible quantity of self-aggregates was found in heart and lung, whereas a small amount (3.6-3.8% of dose) was detected in liver for 3 days after intravenous injection of self-aggregates. The distributed amount of self-aggregates gradually increased in tumor as blood circulation time increased. The concentration of self-aggregates in blood was as high as 14% of dose at 1 day after intravenous injection and was still higher than 8% even at 3 days. When self-aggregates loaded with doxorubicin were administered into the tumor-bearing mice via the tail vein, they exhibited lower toxicity than but comparable anti-tumor activity to free doxorubicin. These results revealed the promising potential of self-aggregates on the basis of glycol chitosan as a carrier for hydrophobic anti-tumor agents.


Asunto(s)
Antineoplásicos/farmacología , Quitosano/química , Doxorrubicina/química , Nanoestructuras/química , Animales , Materiales Biocompatibles/química , Peso Corporal , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Fluoresceína-5-Isotiocianato/farmacología , Pulmón/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Miocardio/metabolismo , Trasplante de Neoplasias , Porcinos , Factores de Tiempo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...