Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nanomedicine ; 56: 102726, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052371

RESUMEN

The pressing demand for innovative approaches to create delivery systems with heightened drug loading and prolonged circulation has spurred numerous efforts, yielding some successes but accompanied by constraints. Our study proposes employing dendritic lipopeptide with precisely balanced opposing charges to extend blood residency for biomimetic nanoplatforms. Neutrally mixed-charged zwitterionic nanoparticles (NNPs) achieved a notable 19 % simvastatin loading content and kept stable even after one-month storage at 4 °C. These nanoplatforms demonstrated low cytotoxicity in NIH-3T3 and L02 cells and negligible hemolysis (<5 %). NNPs inhibited protein adhesion (>95 %) from positively and negatively charged sources through surface hydration. In comparison to positively charged CNPs, NNPs demonstrated an 86 % decrease in phagocytic rate by BMDMs, highlighting their efficacy. Importantly, NNPs showed prolonged circulation compared to CNPs and free simvastatin. These findings highlight the potential of this biomimetic nanoplatform for future therapeutic applications with enhanced drug loading and circulation traits.


Asunto(s)
Biomimética , Nanopartículas , Preparaciones Farmacéuticas , Simvastatina/farmacología , Nanopartículas/química , Sistemas de Liberación de Medicamentos
2.
J Liposome Res ; 34(1): 97-112, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37401372

RESUMEN

To develop a PEGylated and CD44-targeted liposomes, enabled by surface coating with hyaluronic acid (HA) via amide bond to improve the efficacy of imatinib mesylate (IM), for tumor-targeted cytoplasmic drug delivery. HA was covalently grafted on DSPE-PEG2000-NH2 polymer. HA-modified or unmodified PEGylated liposomes were prepared with ethanol injection method, and the stability, drug release, and cytotoxicity of these liposomes were studied. Meanwhile, intracellular drug delivery efficiency, antitumor efficacy, and pharmacokinetics were also investigated. Ex vivo fluorescence biodistribution was also detected by small animal imaging. In addition, endocytosis mechanism was also explored HA-coated PEGylated liposomes (137.5 nm ± 10.24) had a negative zeta potential (-29.3 mV ± 5.44) and high drug loading (27.8%, w/w). The liposomes were stable with cumulative drug leakage (<60%) under physiological conditions. Blank liposomes were nontoxic to Gist882 cells, and IM-loaded liposomes had higher cytotoxicity to Gist882 cells. HA-modified PEGylated liposomes were internalized more effectively than non-HA coating via CD44-mediated endocytosis. Besides, the cellular uptake of HA-modified liposomes also partly depends on caveolin-medicated endocytosis and micropinocytosis. In rats, both liposomes produced a prolonged half-life of IM (HA/Lp/IM: 14.97h; Lp/IM: 11.15h) by 3- to 4.5-folds compared with the IM solution (3.61h). HA-decorated PEGylated liposomes encapsulated IM exhibited strong inhibitory effect on tumor growth in Gist882 cell-bearing nude mice and formation of 2D/3D tumor spheroids. The Ki67 immunohistochemistry result was consistent with the above results. IM-loaded PEGylated liposomes modified with HA exerted the excellent anti-tumor effect on tumor-bearing mice and more drugs accumulated into the tumor site.


Asunto(s)
Ácido Hialurónico , Liposomas , Animales , Ratones , Ratas , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ácido Hialurónico/química , Mesilato de Imatinib/farmacología , Liposomas/química , Ratones Desnudos , Polietilenglicoles/química , Distribución Tisular , Humanos
3.
J Nanobiotechnology ; 19(1): 311, 2021 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-34627266

RESUMEN

Triple-negative breast cancer (TNBC), a management of aggressive breast cancer, remains an unmet medical challenge. Although a wave of efforts had spurred to design novel therapeutic method of TNBC, unpredictable prognosis with lacking effective therapeutic targets along with the resistance to apoptosis seriously limited survival benefits. Ferroptosis is a non-apoptotic form of cell death that is induced by excessive lipid peroxidation, which provide an innovative way to combat cancer. Emerging evidence suggests that ferroptosis plays an important role in the treatment of TNBC cells. Herein, a novel ferroptosis nanomedicine was prepared by loading simvastatin (SIM), a ferroptosis drug, into zwitterionic polymer coated magnetic nanoparticles (Fe3O4@PCBMA) to improve the therapeutic effect of TNBC. The as-obtained Fe3O4@PCBMA-SIM nanoparticles demonstrated more cytotoxicity against MDA-MB-231 than MCF-7 due to the higher expression of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), which demonstrated that statins could effectively kill TNBC. Further experiments showed that SIM could inhibit the expression of HMGCR to downregulate the mevalonate (MVA) pathway and glutathione peroxidase 4 (GPX4), thereby inducing cancer cell ferroptosis. What's more, PCBMA endows Fe3O4@PCBMA longer blood circulation performance to enhance their accumulation at tumor sites. Given that Fe3O4 have proven for clinical applications by the U.S. Food and Drug Administration (FDA) and SIM could induce cancer cell ferroptosis, the developed Fe3O4@PCBMA-SIM nanosystem would have great potential in clinics for overcoming the drug resistance brought about by apoptotic drugs to cancer cells.


Asunto(s)
Ferroptosis/efectos de los fármacos , Simvastatina , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular Tumoral , Preparaciones de Acción Retardada , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Femenino , Humanos , Células MCF-7 , Nanopartículas de Magnetita/química , Masculino , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Simvastatina/química , Simvastatina/farmacocinética , Simvastatina/farmacología
4.
Pharm Dev Technol ; 26(1): 81-91, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33070668

RESUMEN

Nanostructured lipid carriers (NLC) have become a research hotspot, wherein cancer-targeting effects are enhanced and side effects of chemotherapy are overcome. Usually, accelerated blood clearance (ABC) occurs after repeated injections, without changing the immunologic profile, despite PEGylation which prolongs the circulation function. To overcome these problems, we designed a red blood cell-membrane-coated NLC (RBCm-NLC), which was round-like, with a particle size of 60.33 ± 3.04 nm and a core-shell structure. Its stability was good, the drug paclitaxel (PTX) release from RBCm-PTX-NLC was less than 30% at pH7.4 and pH6.5, and the integrity of RBC membrane surface protein was maintained before and after preparation. Additionally, in vitro assays showed that, with the RBCm coating, the cellular uptake of the NLC by cancer cells was significantly enhanced. RBCm-NLC can avoid recognition by macrophage cells and prolong circulation time in vivo. In S180 tumor-bearing mice, the DiR-labeled RBCm-NLC group showed a stronger fluorescence signal and longer retention in tumor tissues, indicating a prompt tumor-targeting effect and extended blood circulation. Importantly, RBCm-PTX-NLC enhanced the antitumor effect and extended the survival period significantly in vivo. In summary, biomimetic NLC offered a novel strategy for drug delivery in cancer therapy.


Asunto(s)
Antineoplásicos/síntesis química , Materiales Biomiméticos/síntesis química , Biomimética/métodos , Portadores de Fármacos/síntesis química , Nanoestructuras/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Femenino , Lípidos , Masculino , Ratones , Nanoestructuras/administración & dosificación , Células RAW 264.7 , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Mol Pharm ; 16(7): 3208-3220, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31145853

RESUMEN

Conventional systemic chemotherapy leads to poor therapeutic outcomes at moments in cancer therapy because the nontargeting anticancer drug release results in adverse effects and consequently drug resistance. The combination therapeutic strategy provides an alternative way to solve the conundrums. Herein, drug delivery systems with a rational design and tumor-targeting abilities become the ideal carriers for combinatorial therapy. IR780 iodide possesses near-infrared fluorescence intensity for fluorescence imaging (FI) and photothermal conversion for photoacoustic imaging (PAI), which also can be employed for tumor phototherapy (including photothermal therapy and photodynamic therapy). However, hydrophobicity and rapid elimination in vivo limit its biomedical applications. Furthermore, the hydrophobicity and high crystallization of IR780 result in poor drug-loading capacity and low stability. In this study, the high-pressure homogenization method was utilized for hydrophobic molecular IR780 and DTX coloading to construct IR780/DTX-PCEC nanoparticles which exhibit narrow size distribution and satisfactory drug-loading capacity. With further erythrocyte membrane [red blood cell (RBC)] camouflaging, the obtained IR780/DTX-PCEC@RBC nanoparticles present desired stability and prolonged circulation time in vivo. Additionally, the IR780/DTX-PCEC@RBC nanoparticles not only can be employed as a FI/PAI dual model imaging probe but also exhibit the property for phototherapy and chemotherapy of tumors. Based on the therapeutic outcome of combination therapy, the IR780/DTX-PCEC@RBC nanoparticles can serve as promising FI- and PAI-guided photo-chemo combination therapy agents for the future treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Digitoxigenina/química , Sistemas de Liberación de Medicamentos/métodos , Membrana Eritrocítica/química , Indoles/química , Nanopartículas/química , Fotoquimioterapia/métodos , Polímeros/química , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Digitoxigenina/farmacocinética , Liberación de Fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Óptica , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Small ; : e1801865, 2018 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-30035856

RESUMEN

Confinement of urate oxidase with detoxifying enzymes into multienzyme architecture is an appealing approach for gout treatment due to its capability to decompose serum uric acid without generation of H2 O2 . However, most of these strategies involve chemical modifications to the enzymes and barely consider enhancing the stability of the multienzyme architectures particularly against proteolysis, which significantly dampened its catalytic activity and in vivo stability. Herein, a novel strategy to prepare multienzyme nanoclusters with highly uricolytic activity and enhanced stability is demonstrated. With the close proximation, catalase can effectively decompose the H2 O2 generated by uricase during uricolysis. Moreover, with a shell structure constructed with polyethylene glycol, the nanocluster achieves great performance in reducing the nonspecific serum protein adsorptions and proteases digestion, leading to an enhanced circulation time after the intravenous administration. Such complementary multienzyme nanoclusters realize the long-term therapeutic effect in the management of serum uric acid level, without any toxicity or undesired immune responses in vivo. This work mimics the synergistic effect of protein complex in nature and can be further developed to a general method for the construction of multienzyme nanoclusters, which provides new opportunities for utilizing therapeutic enzymes for the treatment of metabolic diseases.

7.
Mol Pharm ; 15(8): 3167-3176, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29897762

RESUMEN

Methionine (Met) is one of the essential amino acids of which the transport system L is overexpressed in various tumor cells. In this study, a near-infrared fluorescent dye (IR-780) and methionine were conjugated through a piperazin-polyamines linker to form Cy-Met. The successful synthesis of Cy-Met was validated by optical characterization, NMR, and MS spectra. The absorption peak of Cy-Met was at 680 nm, and the fluorescence peak was at 790 nm. The cytotoxicity assay and cell imaging studies indicated that Cy-Met had good biocompatibility and specific affinity to tumor cells. The dynamic distribution and clearance investigations showed that Cy-Met was eliminated by the liver-intestine pathway. Notably, Cy-Met displayed tumor-targeting ability in U87, H22, and EAC tumor-bearing mice with an evident long circulation time. The results implied that Cy-Met could act as a promising fluorescence probe specialized for long-term tumor monitoring.


Asunto(s)
Colorantes Fluorescentes/administración & dosificación , Metionina/administración & dosificación , Sondas Moleculares/administración & dosificación , Neoplasias/diagnóstico por imagen , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/administración & dosificación , Indoles/química , Microscopía Intravital/métodos , Ensayo de Materiales , Metionina/química , Metionina/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Microscopía Confocal , Microscopía Fluorescente , Sondas Moleculares/química , Neoplasias/patología , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Ultrasound Med ; 37(5): 1243-1256, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29127707

RESUMEN

OBJECTIVES: A long-circulating lipid-coated ultrasound (US) contrast agent was fabricated to achieve a longer wash-out time and gain more resistance against higher-mechanical index sonication. Systemic physical, acoustic, and in vivo imaging experiments were performed to better understand the underlying mechanism enabling the improvement of contrast agent performance by adjusting the physical and acoustic properties of contrast agent microbubbles. METHODS: By simply altering the gas core, a kind of US contrast agent microbubble was synthesized with a similar lipid-coating shell as SonoVue microbubbles (Bracco SpA, Milan, Italy) to achieve a longer wash-out time and higher inertial cavitation threshold. To bridge the structure-performance relationship of the synthesized microbubbles, the imaging performance of the microbubbles was assessed in vivo with SonoVue as a control group. The size distribution and inertial cavitation threshold of the synthesized microbubbles were characterized, and the shell parameters of the microbubbles were determined by acoustic attenuation measurements. All of the measurements were compared with SonoVue microbubbles. RESULTS: The synthesized microbubbles had a spherical shape, a smooth, consistent membrane, and a uniform distribution, with an average diameter of 1.484 µm. According to the measured attenuation curve, the synthesized microbubbles resonated at around 2.8 MHz. Although the bubble's shell elasticity (0.2 ± 0.09 N/m) was comparable with SonoVue, it had relatively greater viscosity and inertial cavitation because of the different gas core. Imaging studies showed that the synthesized microbubbles had a longer circulation time and a better chance of fighting against rapid collapse than SonoVue. CONCLUSIONS: Nano/micrometer long-circulating lipid-coated microbubbles could be fabricated by simply altering the core composition of SonoVue microbubbles with a higher-molecular weight gas. The smaller diameter and higher inertial cavitation threshold of the synthesized microbubbles might make it easier to access deep-seated organs and give prolonged imaging enhancement in the liver.


Asunto(s)
Medios de Contraste/farmacocinética , Aumento de la Imagen/métodos , Lípidos , Neoplasias Hepáticas Experimentales/diagnóstico por imagen , Microburbujas , Ultrasonografía/métodos , Acústica , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Fosfolípidos/farmacocinética , Ratas , Hexafluoruro de Azufre/farmacocinética , Transductores
9.
Mol Pharm ; 14(7): 2236-2244, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28506066

RESUMEN

The overexpression of VEGFR-3 is correlated with a worse prognosis in lung cancer and has been regarded as a rational target for specific drug delivery. Here, VEGFR-3 homing peptide library was efficiently established by computational design. Strong fluorescent signals of selected peptides were observed in A549 cells, but much weaker in other cells. The positive immunostaining overlapped with VEGFR-3 confirmed high affinity and selectivity of one novel peptide (CP-7). In addition, cell uptake of FITC-CP-7 peptide was significantly blocked by coinjection of excess CP-7 peptide. After labeled with 131I, the profile of pharmacology and biodistribution could be traced in vivo. The 131I-radiolabeled CP-7 peptide conjugates were >85% stable in serum over 4 h and exhibited a specific uptake of 18.04 ± 2.04% ID/g at 0.5 h after injection to high VEGFR-3 expressing A549 tumor mice. More importantly, lower uptake concentration in heart (1.06 ± 0.15% ID/g) after 2 h demonstrated the safety of peptide in vivo. The high uptake in the kidneys revealed that renal clearance was the main route of 131I-CP-7 peptide elimination from the body. Lower accumulation of 131I-CP-7 peptide in VEGFR-3 negative HeLa tumor mice further indicated that CP-7 peptide exhibited a higher tumor-homing efficiency. These studies provided a straightforward analytical access to design and screen bioactive peptide based on protein structure and revealed that CP-7 peptide represented a promising homing peptide of VEGFR-3-positive cancer in vitro and in vivo which could be used as a novel target molecule to achieve efficient drug delivery.


Asunto(s)
Péptidos/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química , Células A549 , Animales , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Riñón/metabolismo , Ligandos , Ratones , Péptidos/metabolismo , Radioisótopos/metabolismo , Ratas , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
J Liposome Res ; 27(3): 221-227, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27572825

RESUMEN

Phospholipids and cholesterols are being spotlighted as raw materials for preparing liposomes, one of the key compounds for drug delivery systems (DDS), and as base compounds for converting water-soluble drugs to fat-soluble drugs. Other applications of phospholipids also are being explored. Nippon Fine Chemical, aware of the future of such lipids, has developed new processes for synthesizing and purifying phospholipids and is supplying them on an industrial scale. These products - used worldwide - are highly regarded as raw materials for preparing liposomes. In particular, Nippon Fine Chemical's innovative research led to the development of "Presome®", a base agent that facilitates the preparation of liposome solutions. To further this research, Nippon Fine Chemical has established an "Advanced Lipid Technology".


Asunto(s)
Colesterol/química , Liposomas/química , Fosfolípidos/química , Animales , Preparaciones de Acción Retardada , Industria Farmacéutica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Solubilidad
11.
Drug Dev Ind Pharm ; 43(4): 652-660, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28005445

RESUMEN

In order to improve the in vivo safety and specific delivery efficiency of the antileukemic homoharringtonine (HHT) at the targets, the long-circulating PEGylated liposomes loaded with HHT (LCLipo-HHT) were prepared. Their physical characteristics, in vitro drug release, in vivo pharmacokinetic properties and elementary toxicity were evaluated. The mean diameter of the prepared LCLipo-HHT is 75.6 ± 3.2 nm and the zeta potential is -16.9 ± 2.5 mV. The entrapment efficiency of HHT in the liposomes is 69.5 ± 1.7%. In pharmacokinetic experiments, an increased plasma concentration as well as blood circulation time was obtained when distearoyl phosphoethanolamine-PEG 2000 lipid was added in the formulation, which results in enhancing drug delivery efficiency. Hemolysis test, vascular irritation test and acute toxicity test were used to demonstrate toxicity of LCLipo-HHT. Compared with clinical HHT injection dosage, LCLipo-HHT indicated no vascular irritation, good hemocompatibility, as well as much better safety. Therefore, the prepared LCLipo-HHT can be used as a promising anticancer formulation for antileukemic therapy in the future.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Harringtoninas/química , Harringtoninas/farmacología , Leucemia/tratamiento farmacológico , Liposomas/química , Polietilenglicoles/química , Animales , Química Farmacéutica/métodos , Sistemas de Liberación de Medicamentos/métodos , Homoharringtonina , Ratones , Tamaño de la Partícula , Fosfatidiletanolaminas/química , Conejos
12.
Drug Dev Ind Pharm ; 43(10): 1610-1618, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28481661

RESUMEN

Furanodiene (FN) loaded FA-PEG2000-DSPE modified nanostructured lipid carriers (FA-FN-NLCs) were developed to increase the solubility and bioavailability of FN, prolong the circulation time in blood and improve the targeting ability. FA-FN-NLCs were prepared using emulsification-ultrasonic and low temperature-solidification method and optimized by central composition design (CCD). In vitro and in vivo characteristics of FA-FN-NLCs were investigated in detail. The optimized formulations exhibited a spherical shape with particle size of 127.4 ± 2.62 nm, PDI of 0.268 ± 0.04, zeta potential of -14.7 ± 1.08 mV, high encapsulation efficiency of 89.04 ± 2.26% and loading capacity of 8.46 ± 0.20%. Differential scanning calorimetry (DSC) indicated that FN was not in crystalline state in FA-FN-NLCs. In vitro drug release exhibited a biphasic release pattern which showed a relative burst drug release at the initial time and followed by a prolonged drug release. In vivo, compared with FN solution (FN-SOL) and FN loaded traditional NLCs (FN-NLCs), FA-FN-NLCs had a longer blood circulating time (t1/2) and higher area under the curve (AUC). NiR fluorescence imaging study demonstrated that FA-FN-NLCs specially accumulated in tumor site by the receptor-mediated endocytosis. This study showed that FA-FN-NLCs was a promising drug delivery system for FN in the treatment of cancer.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Ácido Fólico/química , Furanos/química , Compuestos Heterocíclicos con 2 Anillos/química , Lípidos/química , Nanoestructuras/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Disponibilidad Biológica , Rastreo Diferencial de Calorimetría , Química Farmacéutica , Liberación de Fármacos , Furanos/administración & dosificación , Compuestos Heterocíclicos con 2 Anillos/administración & dosificación , Fosfatidiletanolaminas/administración & dosificación , Polietilenglicoles/administración & dosificación , Solubilidad
13.
Pharm Res ; 33(7): 1628-37, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26964546

RESUMEN

PURPOSES: To develop pH-sensitive liposomes (PSL) containing a high content of gemcitabine; and to investigate whether drug loading (DL) would alter the in vitro and pharmacokinetic properties. METHODS: PSL with a high DL were obtained using a modified small-volume incubation method. The DL effects on drug release rate and in vitro cytotoxicity of PSL were evaluated using MIA PaCa-2 pancreatic cancer cells and their pharmacokinetics investigated in rats. RESULTS: The highest DL of 4.5 ± 0.1% was achieved for gemcitabine in PSL with 145 ± 5 nm diameter. DL did not alter the in vitro release rate from PSL. The IC50 (48 h) of PSL (DL 0.5 and 4.5%) and non pH-sensitive liposomes (NPSL, DL 4.2%) were 1.1 ± 0.1, 0.7 ± 0.1 and 37.0 ± 7.5 µM, respectively. The PSL resulted in a 4.2-fold increase in its elimination half-life (6.2 h) compared to gemcitabine solution (1.4 h) in rats. No significant difference in pharmacokinetic parameters was observed between the two PSL (DL 0.5 and 4.5%). CONCLUSION: The PSL offered advantages over NPSL in restoring the sensitivity of pancreatic cancer cells to gemcitabine without requiring a high DL. DL in the PSL did not alter release rate, cytotoxicity or their long-circulating properties. Graphical Abstract ᅟ.


Asunto(s)
Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Liposomas/química , Liposomas/farmacología , Páncreas/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Desoxicitidina/química , Desoxicitidina/farmacología , Liberación de Fármacos/fisiología , Semivida , Concentración de Iones de Hidrógeno , Tamaño de la Partícula , Soluciones Farmacéuticas/química , Soluciones Farmacéuticas/farmacología , Ratas , Ratas Sprague-Dawley , Gemcitabina
14.
Small ; 11(46): 6225-36, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26488923

RESUMEN

For decades, poly(ethylene glycol) (PEG) has been widely incorporated into nanoparticles for evading immune clearance and improving the systematic circulation time. However, recent studies have reported a phenomenon known as "accelerated blood clearance (ABC)" where a second dose of PEGylated nanomaterials is rapidly cleared when given several days after the first dose. Herein, we demonstrate that natural red blood cell (RBC) membrane is a superior alternative to PEG. Biomimetic RBC membrane-coated Fe(3)O(4) nanoparticles (Fe(3)O(4) @RBC NPs) rely on CD47, which is a "don't eat me" marker on the RBC surface, to escape immune clearance through interactions with the signal regulatory protein-alpha (SIRP-α) receptor. Fe(3)O(4) @RBC NPs exhibit extended circulation time and show little change between the first and second doses, with no ABC suffered. In addition, the administration of Fe(3)O(4) @RBC NPs does not elicit immune responses on neither the cellular level (myeloid-derived suppressor cells (MDSCs)) nor the humoral level (immunoglobulin M and G (IgM and IgG)). Finally, the in vivo toxicity of these cell membrane-camouflaged nanoparticles is systematically investigated by blood biochemistry, hematology testing, and histology analysis. These findings are significant advancements toward solving the long-existing clinical challenges of developing biomaterials that are able to resist both immune response and rapid clearance.


Asunto(s)
Materiales Biomiméticos/farmacología , Circulación Sanguínea/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Membrana Eritrocítica/metabolismo , Nanopartículas/química , Animales , Compuestos Férricos/química , Hidrodinámica , Evasión Inmune , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ensayo de Materiales , Ratones , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Polietilenglicoles/química , Células RAW 264.7 , Electricidad Estática , Factores de Tiempo , Distribución Tisular/efectos de los fármacos
15.
Int J Pharm ; 655: 124032, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38521374

RESUMEN

Ferroptosis inhibits tumor growth by iron-dependently accumulating lipid peroxides (LPO) to a lethal extent, which can result from iron overload and glutathione peroxidase 4 (GPX4) inactivation. In this study, we developed biodegradable zwitterionic polymer-cloaked atorvastatin (ATV)-loaded ferric metal-organic frameworks (Fe-MOFs) for cancer treatment. Fe-MOFs served as nanoplatforms to co-deliver ferrous ions and ATV to cancer cells; the zwitterionic polymer membrane extended the circulation time of the nanoparticles and increased their accumulation at tumor sites. In cancer cells, the structure of the Fe-MOFs collapsed in the presence of glutathione (GSH), leading to the depletion of GSH and the release of ATV and Fe2+. The released ATV decreased mevalonate biosynthesis and GSH, resulting in GPX4 attenuation. A large number of reactive oxygen species were generated by the Fe2+-triggered Fenton reaction. This synergistic effect ultimately contributed to a lethal accumulation of LPO, causing cancer cell death. The findings both in vitro and in vivo suggested that this ferroptosis-inducing nanoplatform exhibited enhanced anticancer efficacy and preferable biocompatibility, which could provide a feasible strategy for anticancer therapy.


Asunto(s)
Ferroptosis , Estructuras Metalorgánicas , Neoplasias , Humanos , Polímeros , Atorvastatina , Glutatión , Hierro , Peróxidos Lipídicos , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
16.
J Control Release ; 369: 658-667, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38604384

RESUMEN

Granzyme B (GrB)-based immunotherapy is of interest for cancer treatment. However, insufficient cellular uptake and a lack of targeting remain challenges to make use of GrB for solid tumour therapy. As GrB induced cell death requires the help of perforin (PFN), we designed a system (nGPM) for the co-delivery of GrB and PFN. Therefore, GrB and PFN were loaded in a porous polymeric nanocapsule rich in acetylcholine analogues and matrix metalloproteinase-2 (MMP-2) responsive peptides. The neutrally charged nGPM nanocapsules showed as long circulating time and accumulated at the tumour sites. Once in the tumour the outside shell of nanocapsules became degraded by overexpressed MMP-2 proteases, resulting in the release of GrB and PFN. We found that the PFN complex formed small pores on the surface of tumour cells which allow GrB to enter the cytoplasm of tumour cells inducing cell apoptosis and tumour suppression significantly.


Asunto(s)
Granzimas , Nanocápsulas , Perforina , Granzimas/metabolismo , Nanocápsulas/química , Animales , Perforina/metabolismo , Humanos , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/inmunología , Apoptosis/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Inmunoterapia/métodos , Ratones Endogámicos BALB C , Femenino , Ratones
17.
Nanomaterials (Basel) ; 14(5)2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38470744

RESUMEN

Recent strides in nanomaterials science have paved the way for the creation of reliable, effective, highly accurate, and user-friendly biomedical systems. Pioneering the integration of natural cell membranes into sophisticated nanocarrier architectures, cell membrane camouflage has emerged as a transformative approach for regulated drug delivery, offering the benefits of minimal immunogenicity coupled with active targeting capabilities. Nevertheless, the utility of nanomaterials with such camouflage is curtailed by challenges like suboptimal targeting precision and lackluster therapeutic efficacy. Tailored cell membrane engineering stands at the forefront of biomedicine, equipping nanoplatforms with the capacity to conduct more complex operations. This review commences with an examination of prevailing methodologies in cell membrane engineering, spotlighting strategies such as direct chemical modification, lipid insertion, membrane hybridization, metabolic glycan labeling, and genetic engineering. Following this, an evaluation of the unique attributes of various nanomaterials is presented, delivering an in-depth scrutiny of the substantial advancements and applications driven by cutting-edge engineered cell membrane camouflage. The discourse culminates by recapitulating the salient influence of engineered cell membrane camouflage within nanomaterial applications and prognosticates its seminal role in transformative healthcare technologies. It is envisaged that the insights offered herein will catalyze novel avenues for the innovation and refinement of engineered cell membrane camouflaged nanotechnologies.

18.
Int J Nanomedicine ; 18: 5159-5181, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37705869

RESUMEN

Background: Flurbiprofen axetil (FA) is a non-steroidal anti-inflammatory drug with good analgesic and anti-inflammatory effects. However, it suffers from poor solubility, short circulation time, and off-target binding profile, which significantly limit its clinical application. Here, we loaded FA into stealth lipid microspheres modified with the arginine-glycine-aspartic acid (RGD) peptide (cRGD-FA-SLM), and examined the therapeutic potential of the resulting platform for the treatment of rheumatoid arthritis (RA). Methods: cRGD-FA-SLM was prepared by high pressure homogenization, and its toxicity and uptake by macrophages were examined using cultures of RAW264.7 cells. Hemolysis and hepatotoxicity tests were performed to assess the safety of the developed platform, while its pharmacokinetics, biodistribution, and therapeutic efficacy were investigated in a collagen-induced arthritis rat model. Results: cRGD-FA-SLM showed homogeneous spherical morphology and efficient encapsulation of FA. The developed platform was non-toxic to normal macrophages and was selectively internalized by lipopolysaccharide-activated macrophages in vitro, while it distributed mainly to arthritic joints and significantly prolonged FA in circulation in vivo. cRGD-FA-SLM also significantly reduced the expression of prostaglandin E2 and alleviated joint edema and bone erosion, showing prolonged analgesic effects in arthritic rats. Conclusion: cRGD-FA-SLM shows good inflammation-targeting ability and prolongs drug circulation in vivo, suggesting promise as an anti-inflammatory and analgesic agent for targeted RA treatment.


Asunto(s)
Artritis Reumatoide , Nanosferas , Animales , Ratas , Distribución Tisular , Artritis Reumatoide/tratamiento farmacológico , Dinoprostona
19.
Artículo en Inglés | MEDLINE | ID: mdl-36751121

RESUMEN

RNA interference (RNAi) mediated by short interfering RNA (siRNA) is a promising method for cancer treatment, but the clinical application is hampered by several limitations, including metabolic instability, lack of tumor specificity, and poor cellular uptake. To meet these challenges, we have explored the possibility of structure modification of siRNA with artificial bases for property optimization. A series of siRNAs functionalized with different numbers of hydrophobic base F are prepared for screening. The interactions of plasma proteins with F-base-modified siRNA (F-siRNA) are investigated, and it is identified that the interaction with serum albumin is dominant. Experiments revealed that the introduction of F bases conferred modified siRNA with improved tumor-specific accumulation, prolonged circulatory retention time, and better tissue permeability. Mechanistic studies indicated that the F base induces the formulation of a stable siRNA-albumin complex, which transports siRNA to tumor tissues selectively owing to an enhanced permeability and retention (EPR) effect of albumin. The F base also facilitates the binding of siRNA to transport-associated proteins on the cell membrane, enabling its cellular internalization. Together, these data demonstrate that F base modification confers siRNA-enhanced cellular uptake and biostability and specific accumulation in tumor tissue, which provides a new approach for the development of siRNA-based cancer therapeutics.

20.
J Control Release ; 364: 23-36, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37863358

RESUMEN

PEGylated cholesterol-containing liposomes (Chol-PEG-lipo) have been widely used as a drug carrier for their good stealth property in blood circulation where cholesterol maintains the stability of the liposomal lipid bilayer and PEGylation endows liposomes with long circulation capability. However, cholesterol-related disadvantages and the accelerated blood clearance (ABC) phenomenon caused by PEGylation greatly limit the application of conventional stealth liposomes in clinic. Herein, ginsenoside Rg3 was selected to substitute cholesterol and PEG for liposomes preparation (Rg3-lipo). Rg3 was proved with similar liposomal membrane regulation ability to cholesterol and comparable long circulation effect to PEG. In addition, repeated administrations of Chol-PEG-lipo and Rg3-lipo were performed. The circulation time of the second dose of Chol-PEG-lipo was substantially reduced accompanied by a greatly increased accumulation in the liver due to the induction of anti-PEG IgM and the subsequent activated complement system. In contrast, no significantly increased level of relative plasma cells, IgM secretion and the complement activation in blood circulation was observed after the second injection of Rg3-lipo. As a result, Rg3-lipo showed great stealth property without ABC phenomenon. Therefore, developing liposomes utilizing Rg3 instead of PEG and cholesterol presents a promising strategy to prolong the blood circulation time of liposomes without triggering the ABC phenomenon and activated immune responses.


Asunto(s)
Liposomas , Polietilenglicoles , Ratas , Animales , Ratas Wistar , Inmunoglobulina M , Colesterol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA