Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Nanomedicine ; 19: 189-208, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38223882

RESUMEN

Purpose: Liver cancer is considered as the third leading cause of cancer-related deaths, with hepatocellular carcinoma (HCC) accounting for approximately 90% of liver cancers. Improving the treatment of HCC is a serious challenge today. The primary objective of this study was to construct SP94-Fe3O4@ICG&DOX nanoparticles and investigate their potential diagnosis and treatment effect benefits on HCC. Methods: Firstly, we synthesized and characterized SP94-Fe3O4@ICG&DOX nanoparticles and confirmed their in vitro release behavior, photothermal and photodynamic performance. Moreover, the in vivo imaging capability was also observed. Finally, the inhibitory effects on Hepa1-6 in vitro and in vivo were observed as well as biosafety. Results: SP94-Fe3O4@ICG&DOX nanoparticles have a size of ~22.1 nm, with an encapsulation efficiency of 45.2% for ICG and 42.7% for DOX, showing excellent in vivo MPI and fluorescence imaging capabilities for precise tumor localization, and synergistic photo-chemotherapy (pH- and thermal-sensitive drug release) against tumors under irradiation. With the assistance of a fluorescence molecular imaging system or MPI scanner, the location and contours of the tumor were clearly visible. Under a constant laser irradiation (808 nm, 0.6 W/cm2) and a set concentration (50 µg/mL), the temperature of the solution could rapidly increase to ~45 °C, which could effectively kill the tumor cells. It could be effectively uptaken by HCC cells and significantly inhibit their proliferation under the laser irradiation (100% inhibition rate for HCC tumors). And most importantly, our nanoparticles exhibited favorable biocompatibility with normal tissues and cells. Conclusion: This versatile agent can serve as an intelligent and promising nanoplatform that integrates multiple accurate diagnoses, precise positioning of cancer tissue, and effective coordination with synergistic tumor photodynamic therapy.


Asunto(s)
Carcinoma Hepatocelular , Hipertermia Inducida , Neoplasias Hepáticas , Nanopartículas , Humanos , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Fototerapia/métodos , Doxorrubicina , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Hipertermia Inducida/métodos , Línea Celular Tumoral
2.
ACS Nano ; 17(15): 14800-14813, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37486924

RESUMEN

The development of functional materials for tumor immunogenicity enhancement is desirable for overcoming the low therapeutic efficiency and easy metastasis during tumor treatments. Herein, the thermoresponsive nanoparticles composed of photothermal agent (PTA) and click reactive reagent are developed for enhanced immunotherapy application. A Ni-bis(dithiolene)-containing PTA with intense near-infrared absorption and efficient photothermal conversion is developed for thermoresponsive nanoparticles construction. The generated heat by encapsulated PTA further induces the phase transition of thermoresponsive nanoparticles with the release of chemotherapy reagent to react with the amino groups on functional proteins, realizing PTT and chemotherapy simultaneously. Moreover, the immunogenic cell death (ICD) of cancer cells evoked by PTT could be further enhanced by the released reactive reagent. As a result, the synergistic effect of photothermal treatment and reaction-mediated chemotherapy can suppress the growth of a primary tumor, and the evoked ICD could further activate the immune response with the suppression of a distant tumor. This synergistic treatment strategy provides a reliable and promising approach for cancer immunotherapy in clinic.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Ratones , Terapia Fototérmica , Fototerapia , Neoplasias/terapia , Antígenos de Neoplasias , Línea Celular Tumoral , Inmunoterapia
3.
ACS Appl Mater Interfaces ; 15(12): 15129-15139, 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-36919267

RESUMEN

Due to the complexity of tumor pathogenesis and the heterogeneity of the tumor microenvironment (TME), it is difficult to obtain satisfactory efficacy with a single therapy. In this study, a hyaluronic acid (HA)-modified ruthenium nanoaggregate (RuNA) and glucose oxidase (GOD) -loaded manganese dioxide (MnO2) nanoflowers (MRG@HA) have been prepared. RuNA and MnO2 nanoflowers can generate O2 in TME, alleviating tumor tissue hypoxia. RuNA is a good photothermal agent for high-temperature ablation of solid tumors under infrared laser irradiation. GOD consumes glucose in the presence of O2 and converts it into glucuronic acid and hydrogen peroxide, reducing tumor nutrient supply while promoting Fenton-like reactions of MnO2 nanoflowers and RuNA to produce cytotoxic hydroxyl radicals. MRG@HA can also actively target tumor cells through the affinity of HA and CD44 receptor to improve the antitumor effect. In vitro and in vivo studies have confirmed the synergistic effect of MRG@HA with tumor photothermal/chemodynamic/starvation therapy, showing its great potential for clinical application in tumor therapy.


Asunto(s)
Terapia por Estimulación Eléctrica , Nanopartículas , Neoplasias , Humanos , Compuestos de Manganeso/farmacología , Óxidos , Neoplasias/tratamiento farmacológico , Manejo del Dolor , Glucosa Oxidasa , Peróxido de Hidrógeno , Microambiente Tumoral , Línea Celular Tumoral
4.
Eur J Pharm Sci ; 180: 106319, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36328086

RESUMEN

Glioblastoma (GBM) is a common malignant tumor in brain, and the treatment is still a challenge owing to the high invasiveness and the existence of blood-brain barrier (BBB). Although temozolomide (TMZ) is the first line medication, its efficacy is not ideal, which is related to the defect of dose distribution and drug resistance. It is urgent to develop a novel BBB-permeable nanoagent with multiple therapeutic modalities for improving the treatment effect of GBM. In this work, we constructed an intelligent BBB-permeable nanoplatform (CTHG-Lf NPs) with hollow mesoporous copper sulfide nanoparticles (HM-CuS NPs) as temozolomide (TMZ) carrier and hyaluronic acid (HA) as gatekeeper, as well as further modification with glucose oxidase (GOx) and lactoferrin (Lf) for highly efficient synergistic therapy of orthotopic GBM. The modification of Lf endows CTHG-Lf NPs with good target and BBB-permeable ability. HA not only prevents the TMZ leakage during circulation, but also achieves responsive drug release at tumor site for effective chemotherapy (CT). GOx provides high hydrogen peroxide (H2O2) and gluconic acid for improving the treatment effect of chemodynamic therapy (CDT), and realizes the starvation therapy (ST) by consuming glucose. The good photothermal effect of CTHG-Lf NPs achieves the "mild" photothermal therapy (PTT), while enhancing the efficiency of Fenton-like reaction. The synergistic strategy with CT/CDT/PTT/ST can not only promote brain drug delivery, but also realize the combination of multiple mechanisms for effective tumor growth suppression in vivo.


Asunto(s)
Glioma , Nanopartículas , Neoplasias , Humanos , Fototerapia , Barrera Hematoencefálica , Terapia Fototérmica , Peróxido de Hidrógeno , Ácido Hialurónico/farmacología , Glioma/tratamiento farmacológico , Neoplasias/patología , Temozolomida , Línea Celular Tumoral
5.
Artículo en Chino | WPRIM | ID: wpr-993168

RESUMEN

Thermotherapy has become another important tumor treatment after surgery, radiotherapy, chemotherapy, and targeted treatment. Magnetic hyperthermia (MH) is a new type of hyperthermia, which has attracted widespread attention due to its advantages of non-invasiveness / minimal invasiveness, high efficiency and good tissue penetration. It provides a new option for the molecular level treatment of malignant tumors with high efficacy and low toxicity, which has become a new research direction of tumor treatment. Magnetic materials and suitable magnetic fields are needed to realize MH. Iron oxide nanoparticles (IONs) are widely studied as MH agents because of their high biocompatibility and heating ability. In this article, the research progress on magnetic iron oxide nanomaterials and MH combined with antitumor therapy based on magnetic nanoparticles were analyzed, and the potential application of MH in cancer treatment was reviewed.

6.
J Colloid Interface Sci ; 625: 614-627, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35764043

RESUMEN

A novel nanoplatform that supports multimodal imaging has been designed for deep tumor therapy. In this study, Bi2Se3@Cu2-xSe heterojunction nanocomposites with tunable spectral absorption, effective electron-hole separation and high photothermal conversion efficiency were prepared for the combination therapy of phototherapy (PT), chemodynamic therapy (CDT) and radiotherapy (RT). By adjusting the doping ratio, the heterojunction nanoparticles show obvious tunable ability of local surface plasmon resonance and the ability to promote electron-hole separation with significantly enhanced reactive oxygen species production capacity. The band structure and charge density difference calculated by density functional theory further reveal that the change of band gap and the decrease of free carriers can regulate the spectral absorption of nanomaterials and promote electron-hole separation. In addition, the photothermal conversion properties of low carrier density semiconductors are related to their inherent deep level defects. The formation of heterojunctions making the Se atoms deviate from the Bi2Se3 lattice, resulting in more deep level defects and stronger photothermal conversion properties. Meanwhile, this nanoplatform presented features similar to catalase activities and glutathione (GSH) consumption characteristics, which was capable of effectively alleviate the tumor-specific hypoxia environment to enhance the efficacy of O2-dependent photodynamic therapy (PDT) and radiotherapy (RT) and depletion GSH to prevent the reduction of therapeutic efficacy due to the clearance of reactive oxygen species. In addition to therapeutic enhancement, heterojunction nanomaterials have excellent nuclear magnetic resonance imaging (MRI), infrared thermal imaging (IR) and computed tomography (CT) properties due to their significant paramagnetism and excellent photothermal conversion and X-ray attenuation capacities. In conclusion, our findings provide a new strategy for designing multi-function and efficient nanoplatform to treat tumor.


Asunto(s)
Nanocompuestos , Neoplasias , Fotoquimioterapia , Línea Celular Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Fotoquimioterapia/métodos , Fototerapia/métodos , Especies Reactivas de Oxígeno
7.
J Colloid Interface Sci ; 623: 124-134, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35569221

RESUMEN

Targeting excess H2O2 in the tumor microenvironment, nanotheranostic agents for catalytic therapy are designed based on Fenton reaction, catalyzing H2O2 into oxygen and hydroxyl radical (OH). But the catalytic efficiency in tumor microenvironment is not satisfactory. In order to solve the problem, a series of bimetallic-dual ligands metal-organic frameworks Fe/Tm-MOFs were designed, that Fe3+ and Tm3+ as metalions, 2-methylimidazole and trimesic acid as ligands. Due to the doped Tm3+ in Fe/Tm-MOFs and the conjugated structures formed by two ligands, the rate of electron transfer was improved, thus promoting the generation of OH at some extent. In addition, the photothermal effect of Fe/Tm-MOFs further promotes the generation of OH, which was evidenced by the 3,3',5,5'-tetramethylbenzidine(TMB). Combining the drug loading and release capabilities of Fe/Tm-MOFs, synergetic therapy of photothermal/chemo-/catalytic therapy can be achieved. In vitro results reveal that DOX release behaviors are both pH- and thermal-responsive. In vivo anti-cancer results show that the tumors of mice almost disappeared within 10 days, which were injected with Fe/Tm-MOFs/DOX and irradiated with 808 nm for 10 min. Thus, an excellent therapeutic performance has been achieved. Besides, Fe/Tm-MOFs can serve as a multimodality bioimaging contrast agent, covering fluorescence imaging, photothermal imaging and magnetic resonance imaging. Thus, an all-in-one nanotheranostic agent is constructed, improving the catalytic efficiency and providing a novel method to design an efficient nanotheranostic agent.


Asunto(s)
Hipertermia Inducida , Estructuras Metalorgánicas , Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Peróxido de Hidrógeno , Hipertermia Inducida/métodos , Ligandos , Estructuras Metalorgánicas/química , Ratones , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Fototerapia/métodos , Microambiente Tumoral
8.
Drug Deliv ; 29(1): 1312-1325, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35475384

RESUMEN

Photothermal therapy (PTT) has become one of the most promising therapies in cancer treatment as its noninvasiveness, high selectivity, and favorable compliance in clinic. However, tumor thermotolerance and distal metastasis reduce its efficacy, becoming the bottleneck of applying PTT in clinic. In this study, a chidamide-loaded magnetic polypyrrole nanocomposite (CMPP) has been fabricated as a visualized cancer photothermal agent (PTA) to counter tumor thermotolerance and metastasis. The efficacy of CMPP was characterized by in vitro and in vivo assays. As a result, this kind of magnetic polypyrrole nanocomposites were black spherical nanoparticles, possessing a favorable photothermal effect and the suitable particle size of 176.97 ± 1.45 nm with a chidamide loading rate of 12.92 ± 0.45%. Besides, comparing with PTT, CMPP exhibited significantly higher cytotoxicity and cellular apoptosis rate in two tumor cell lines (B16-F10 and HepG2). In vivo study, the mice showed obvious near-infrared (NIR) and magnetic resonance imaging (MRI) dual-modal imaging at tumor sites and sentinel lymph nodes (SLNs); on the other hand, magnetic targeting guided CMPP achieved a cure level on melanoma-bearing mice through preventing metastasis and thermotolerance. Overall, with high loading efficiency of chidamide and strong magnetic targeting to tumor sites and SLNs, CMPP could significantly raise efficiency of PTT by targeting tumor thermotolerance and metastasis, and this strategy may be exploited therapeutically to upgrade PTT with MPP as one of appropriate carriers for histone deacetylase inhibitors (HDACis).


Asunto(s)
Neoplasias , Termotolerancia , Aminopiridinas , Animales , Benzamidas , Imagen por Resonancia Magnética/métodos , Ratones , Neoplasias/tratamiento farmacológico , Fototerapia , Terapia Fototérmica , Polímeros/química , Pirroles
9.
ACS Nano ; 16(1): 869-884, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34985280

RESUMEN

The treatment of Parkinson's disease (PD) has been hindered by the complex pathologies and multiple membrane barriers during drug delivery. Although exosomes derived from mesenchymal stem cells (MSCs) have great potential for PD, MSC-derived exosomes alone could not fully meet the therapeutic requirements due to their limitation in therapy and delivery. Here, we develop a self-oriented nanocarrier called PR-EXO/PP@Cur that combines therapeutic MSC-derived exosomes with curcumin. PR-EXO/PP@Cur can be self-oriented across the multiple membrane barriers and directly release drugs into the cytoplasm of target cells after intranasal administration. With enhanced accumulation of drugs in the action site, PR-EXO/PP@Cur achieves three-pronged synergistic treatment to deal with the complex pathologies of PD by reducing α-synuclein aggregates, promoting neuron function recovery, and alleviating the neuroinflammation. After treatment with PR-EXO/PP@Cur, the movement and coordination ability of PD model mice are significantly improved. These results show that PR-EXO/PP@Cur has great prospects in treatment of PD or other neurodegenerative diseases.


Asunto(s)
Curcumina , Exosomas , Células Madre Mesenquimatosas , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/tratamiento farmacológico , Administración Intranasal , Curcumina/uso terapéutico
10.
Chemosphere ; 293: 133661, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35063560

RESUMEN

The integrated wastewater discharge standard for phosphorus has become increasingly strict. In this study, a synergetic current stimulation system coupled with anaerobic digestion was used to enhance phosphorus removal from wastewater. The effects of current intensity, pH, and methane (CH4) synthesis on phosphorus removal were investigated. As direct current was supplied to an anaerobic bioreactor, the removal of sewage total phosphorus was significantly enhanced. The conditions of weak acid and low negative oxidation-reduction potential facilitated the phosphorus removal from wastewater. The optimal parameters for the dephosphorisation process were a current intensity of 100 mA and a pH of 6.0. When the anaerobic digestion process was inhibited by the reagent 2-bromoethanesulphonic acid sodium (BES), abundant metabolic intermediates accumulated and methanogenesis clearly decreased. Affected by the current stimulation and the inhibition of CH4 synthesis, the formation of gaseous phosphine (PH3) was greatly improved, and then PH3 escaped from the digestion mixture after it was absorbed by microbial cells. The maximum PH3 content of the digestion gas was 41.8 mg m-3 in the reactor supplied with a current of 100 mA and BES addition of 10 mmol L-1, and the phosphorus removal in this digestion system reached 55.2% at 6 d; however, the removal in the conventional anaerobic digestion system was only 17.7% after the same amount of time. Finally, a pathway of enhanced anaerobic biological phosphorus removal was proposed to better understand the inherent synergistic mechanism.


Asunto(s)
Fósforo , Aguas Residuales , Anaerobiosis , Reactores Biológicos , Fósforo/metabolismo , Aguas del Alcantarillado/química
11.
J Colloid Interface Sci ; 610: 313-320, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-34923269

RESUMEN

Dense tumor stroma is the physiological barrier in drug delivery that prevents anticancer drugs from entering the tumor, thereby seriously limiting the drugs' therapeutic effect. In this study, a Janus nanoplatform consisting of periodic mesoporous organosilica-coated platinum nanoplatforms (JPMO-Pt) and anti-stroma drug halofuginone (HF) (denoted as JPMO-Pt-HF), was developed to deplete the tumor stroma and synergistically treat breast cancer in BALB/c mice. The prepared JPMO-Pt had a uniform size of 245 nm, a good dispersion, an excellent in vitro and in vivo biocompatibility, and a high loading capacity for HF (up to 50 µg/mg). The antitumor experiments showed that the survival rate of 4 T1 cells exhibited an obvious downward trend when the cells were incubated with the JPMO-Pt-HF and irradiated with 808 nm laser. Moreover, the cell survival rate was only about 10% at 48 h when the HF concentration was 2.0 µg/mL. Notably, JPMO-Pt-HF under irradiation had an excellent synergistic therapeutic effect on tumor cells. In vivo antitumor experiment further showed that the JPMO-Pt-HF, in combination with laser irradiation, could minimize tumor growth, showing significantly better effects than those observed for the case of monotherapy involving photothermal therapy (PTT) (152 vs. 670 mm3, p < 0.0001) and HF (152 vs. 419 mm3, p = 0.0208). In addition, immunohistochemistry of tumor tissues indicated that JPMO-Pt-HF obviously reduced the relative collagen and α-smooth muscle actin (α-SMA) area fraction. Taken together, this research designs a new platform that not only possesses the ability to degrade the tumor matrix but also combines PTT and chemotherapeutic effects, and holds promise for effective tumor treatment.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Animales , Línea Celular Tumoral , Doxorrubicina , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Ratones Endogámicos BALB C , Fototerapia , Terapia Fototérmica , Piperidinas , Quinazolinonas
12.
J Colloid Interface Sci ; 605: 296-310, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34329981

RESUMEN

This paper presents the design of a new type of intelligent and versatile all-in-one therapeutic nanoplatform for the co-delivery of chemotherapeutic drugs and photosensitizers to facilitate multimodal antitumor treatment; the system is based on hyaluronic acid (HA)-modified manganese dioxide (MnO2)-enveloped hollow porous copper sulfide (CuS) nanoparticles (CuS@MnO2/HA NPs). In this system, a CuS inner shell allows for the co-loading of doxorubicin (DOX) and indocyanine green (ICG) and induces photothermal effects, and a biodegradable MnO2 external shell affords on-demand tumor microenvironment (TME)-triggered release and catalase- andFenton-like activities. Moreover, the HA modification endows the system with a CD44 receptor-mediated tumor-targeting property. The formulated DOX and ICG co-loaded CuS@MnO2/HA (DOX/ICG-CuS@MnO2/HA) NPs were found to exhibit excellent photothermal performance both in vitro and in vivo. In addition, DOX/ICG-CuS@MnO2/HA NPs were found to display both TME and near-infrared (NIR)-responsive controlled release properties. The NPs also have a superior reactive oxygen species (ROS) generation capacity due to the combination of enhanced ICG-induced singlet oxygen and CuS@MnO2-mediated hydroxyl radicals. The cellular uptake, fluorescence imaging property, cytotoxicity, and thermal imaging of these NPs were also evaluated. In tumor-bearing mice, the DOX/ICG-CuS@MnO2/HA NPs displayeda superior antitumor efficacy (2.57-fold) as compared with free DOX. Therefore, the developed DOX/ICG-CuS@MnO2/HA NPs have a great potential for use as an all-in-one nanotherapeutic agent for the efficient and precise induction of chemo/photothermal/photodynamic/chemodynamic therapy with superior antitumor efficacy and fewer side effects.


Asunto(s)
Nanopartículas , Preparaciones Farmacéuticas , Animales , Cobre , Doxorrubicina/farmacología , Ácido Hialurónico , Compuestos de Manganeso , Ratones , Óxidos , Fármacos Fotosensibilizantes , Fototerapia , Sulfuros
13.
Drug Des Devel Ther ; 15: 4105-4123, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616144

RESUMEN

PURPOSE: The effective treatment of ulcerative colitis (UC) poses substantial challenges, and the aetiopathogenesis of UC is closely related to infectious, immunological and environmental factors. Currently, there is a considerable need for the development of orally bioavailable dosage forms that enable the effective delivery of therapeutic drugs to local diseased lesions in the gastrointestinal tract. METHODS: Berberine (BBR) and Atractylodes macrocephala Koidz (AM) volatile oil, derived from the Chinese herbs Coptis chinensis Franch and Atractylodes macrocephala Koidz, have anti-inflammatory and immunomodulatory activities. In this study, we prepared colon-targeted pellets loaded with BBR and stomach-targeted pellets loaded with AM volatile oil for the synergistic treatment of UC. The Box-Behnken design and ß-cyclodextrin inclusion technique were used to optimize the enteric coating formula and prepare volatile oil inclusion compounds. RESULTS: The two types of pellets were spherical and had satisfactory physical properties. The pharmacokinetic results showed that the AUC and MRT values of the dual-targeted (DPs) pellets were higher than those of the control pellets. In addition, in vivo animal imaging confirmed that the DPs could effectively deliver BBR to the colon. Moreover, compared with sulfasalazine and monotherapy, DPs exerted a more significant anti-inflammatory effect by inhibiting the expression of inflammatory factors including IL-1ß, IL-4, IL-6, TNF-α and MPO both in serum and tissues and enhancing immunity by decreasing the production of IgA and IgG. CONCLUSION: The DPs play a synergistic anti-UC effect by exerting systemic and local anti-inflammatory and provide an effective oral targeted preparation for the treatment of UC.


Asunto(s)
Berberina/farmacología , Colitis Ulcerosa/tratamiento farmacológico , Aceites Volátiles/farmacología , Administración Oral , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Área Bajo la Curva , Atractylodes/química , Berberina/aislamiento & purificación , Berberina/farmacocinética , Química Farmacéutica , Colitis Ulcerosa/fisiopatología , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Masculino , Ratones , Ratones Endogámicos BALB C , Aceites Volátiles/aislamiento & purificación , Aceites Volátiles/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
14.
Int J Pharm ; 607: 120947, 2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34358541

RESUMEN

With ideal optical properties, semiconducting polymer quantum dots (SPs) have become a research focus in recent years; a considerable number of studies have been devoted to the application of SPs in non-invasive and biosafety phototherapy with near-infrared (NIR) lasers. Nevertheless, the relatively poor stability of SPs in vitro and in vivo remains problematic. PCPDTBT was chosen to synthesize photothermal therapy (PTT) and photodynamic therapy (PDT) dual-model SPs, considering its low band gap and desirable absorption in the NIR window. For the first time, cetrimonium bromide was used as a stabilizer to guarantee the in vitro stability of SPs, and as a template to prepare SP hybrid mesoporous silica nanoparticles (SMs) to achieve long-term stability in vivo. The mesoporous structure of SMs was used as a reservoir for the hypoxia-activated prodrug Tirapazamine (TPZ). SMs were decorated with polyethylene glycol-folic acid (SMPFs) to specifically target activated macrophages in rheumatoid arthritis (RA). Upon an 808 nm NIR irradiation, the SMPFs generate intracellular hyperthermia and excessive singlet oxygen. Local hypoxia caused by molecular oxygen consumption simultaneously activates the cytotoxicity of TPZ, which effectively kills activated macrophages and inhibits the progression of arthritis. This triple PTT-PDT-chemo synergistic treatment suggests that SMPFs realize the in vivo application of SPs and may be a potential nano-vehicle for RA therapy with negligible side-toxicity.


Asunto(s)
Artritis Reumatoide , Hipertermia Inducida , Nanopartículas , Fotoquimioterapia , Artritis Reumatoide/tratamiento farmacológico , Ácido Fólico , Humanos , Fototerapia , Terapia Fototérmica , Polímeros , Dióxido de Silicio
15.
Drug Deliv ; 28(1): 171-182, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33427515

RESUMEN

10-HCPT is a topoisomerase I inhibitor effective in the treatment of liver cancer but its use is hampered by its resistance. The expression of hypoxia-inducible factor-1α (HIF-1α) is reportedly upregulated in liver cancer tissues, which is directly linked to the resistance of 10-HCPT. While BBR can significantly decrease the level of HIF-1α according to the literature report. Thus, the aim of this study was to prepare a novel intravenous 10-HCPT-BBR-loaded lipid microsphere (LM) and evaluate their synergistic effect on liver cancer treatment. The optimal preparation mainly included 10.0% oil phase (medium-chain triglyceride:long-chain triglyceride = 1:1), emulsifier (egg lecithin E80 and pluronic F68), antioxidant (0.02% NaHSO3), and pH regulator (0.1 mol/L Hcl). Then, the behaviors of BBR-10-HCPT loaded LM in vitro and in vivo were systematically investigated. In vitro, it showed an obvious sustained-release effect in different release mediums, good physicochemical stability at accelerated and long-term storage conditions, and great anti-proliferative capability toward human liver cancer Hep-3B cells. In vivo, the prepared LM exhibited a longer half-life and higher AUC compared to BBR injection and 10-HCPT injection. More importantly, it was found that The LM was distributed more in the liver, spleen, and tumors, but less in the lungs and heart, especially in the lung. And then, it showed significant inhibition of tumor growth against nude mouse with Hep-3B tumor, and the tumor inhibition rate reached 91.55%. Thus, the data obtained in our study suggested that BBR combined with 10-HCPT can raise curative effect and reduce the toxicity of 10-HCPT.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Berberina/farmacología , Camptotecina/análogos & derivados , Carcinoma Hepatocelular , Neoplasias Hepáticas , Microesferas , Inhibidores de Topoisomerasa I/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Berberina/administración & dosificación , Berberina/química , Camptotecina/administración & dosificación , Camptotecina/química , Camptotecina/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Ratones , Ratones Desnudos , Ratas , Inhibidores de Topoisomerasa I/administración & dosificación , Inhibidores de Topoisomerasa I/química , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Drug Deliv ; 27(1): 983-995, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32611218

RESUMEN

To improve synergistic anticancer efficacy and minimize the adverse effects of chemotherapeutic drugs, temozolomide (TMZ) and curcumin (CUR) co-loaded nanostructured lipid carriers (NLCs) were prepared by microemulsion in this study. And the physicochemical properties, drug release behavior, intracellular uptake efficiency, in vitro and in vivo anticancer effects of TMZ/CUR-NLCs were evaluated. TMZ/CUR-NLCs showed enhanced inhibitory effects on glioma cells compared to single drug loaded NLCs, which may be owing to that the quickly released CUR can sensitize the cancer cells to TMZ. The inhibitory mechanism is a combination of S phase cell cycle arrest associated with induced apoptosis. Notably, TMZ/CUR-NLCs can accumulate at brain and tumor sites effectively and perform a significant synergistic anticancer effect in vivo. More importantly, the toxic effects of TMZ/CUR-NLCs on major organs and normal cells at the same therapeutic dosage were not observed. In conclusion, NLCs are promising nanocarriers for delivering dual chemotherapeutic drugs sequentially, showing potentials in the synergistic treatment of tumors while reducing adverse effects both in vitro and in vivo.


Asunto(s)
Antineoplásicos/farmacología , Curcumina/farmacología , Portadores de Fármacos/química , Nanopartículas/química , Temozolomida/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Química Farmacéutica/métodos , Curcumina/administración & dosificación , Curcumina/farmacocinética , Combinación de Medicamentos , Liberación de Fármacos , Glioma/tratamiento farmacológico , Humanos , Lípidos/química , Tamaño de la Partícula , Fase S/efectos de los fármacos , Temozolomida/administración & dosificación , Temozolomida/farmacocinética
17.
Acta Biomater ; 110: 208-220, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32278084

RESUMEN

The heat shock response (HSR) induced by photothermal therapy (PTT), which can cause tumor cells to resist apoptosis, has increasingly attracted the attention of researchers. Synergistic treatment of tumors using multiple means to improve therapeutic efficiency would be a promising strategy for effective cancer treatment. In this study, a cancer cell membrane-camouflaged nanocarrier was developed and loaded with tellurium (Te) and cantharidin (CTD) for efficient combinatorial therapy. The designed nanoparticles (m-CTD@Te) used a 4T1 cell membrane coating as the shell with homologous targeting capability, CTD as an the HSR inhibitor and antitumor drug, and Te as a PTT and photodynamic therapy (PDT) photosensitizer. An in vivo study indicated that the tumor inhibition rate of this combinatorial therapy could reach approximately 82.3% in 4T1 mammary tumor models. This study suggested that m-CTD@Te, as a versatile biomimetic nanoplatform, provides a new alternative for more precise and effective tumor treatment. STATEMENT OF SIGNIFICANCE: In this work, we constructed cell membrane-coated biomimetic nanoparticles (m-CTD@Te) to suppress cancer effectively through synergistic treatment. The developed m-CTD@Te nanoparticles presented strong homologous targeting capabilities. The encapsulated Te triggered PDT and PPT under the near-infrared laser irradiation. Subsequently, the PTT triggered the release of CTD, which could suppress the HSR of tumor and achieve chemotherapy. In addition, due to the presence of outer cell membrane coating, these m-CTD@Te nanoparticles showed good biocompatibility to healthy cells.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Biomimética , Cantaridina , Respuesta al Choque Térmico , Humanos , Fototerapia , Terapia Fototérmica , Telurio
18.
Colloids Surf B Biointerfaces ; 187: 110631, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31776054

RESUMEN

Cancer is a huge challenge humanity facing today, and single chemical treatments inevitably have shortcomings such as poor selectivity and large side effects. This paper constructed an egg yolk phospholipids modified molybdenum disulfide (MoS2) nanocarrier system for the treatment of tumors via the combination of chemotherapy and photothermal therapy. The lipid-modified layered MoS2 (MoS2-Lipid) nanocomposite was synthesized by simple physical adsorption. The lipid modification strongly enhanced the stability of MoS2 nanosheets and the nanocarrier has a large drug loading amount with pH dependent DOX release profile, an excellent photothermal property, and an ideal cellular uptake property. Therefore, we combined chemotherapy and photothermal therapy to treat tumors synergistically. Through in vitro cell experiments, pure nanocomposite had no obvious cytotoxicity to cells, and the synergistic treatment of tumors by chemotherapy and photothermal therapy was more effective than any single treatment. More importantly, in vivo experiments indicated that lipid modification enhanced the accumulation of the nanocarrier in mice tumors, thus a better photothermal performance could be seen compared with original MoS2 nanosheets. In summary, the MoS2-lipid nanocomposite is a promising nanocarrier for the treatment of tumors by chemo and photothermal therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Materiales Biomiméticos/química , Disulfuros/química , Hipertermia Inducida , Molibdeno/química , Nanopartículas/química , Neoplasias/terapia , Fosfolípidos/química , Fototerapia , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Humanos , Células MCF-7 , Ratones Endogámicos ICR , Nanopartículas/ultraestructura , Albúmina Sérica Bovina/química , Espectroscopía Infrarroja por Transformada de Fourier
19.
Biomaterials ; 216: 119220, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31200144

RESUMEN

In this study, we synthesized a thermosensitive composite of Gel-SOR-LUF-SeNPs to achieve the localized synergistic chemoradiotherapy of hepatocellular carcinoma (HCC). Sorafenib (SOR) is one of the important clinical drugs for unresectable and advanced HCC. However, the uncontrollable release of SOR induced drug resistance and severe side effects. Recently, thermosensitive hydrogels have emerged as promising drug-delivery carriers, due to their superior advantages including biodegradability, low-toxicity, high drug loading, site-specificity, sustained and controlled drug release behavior. We synthesized the thermosensitive hydrogel nanosystem (Gel-SOR-LUF-SeNPs) as an effective drug release depot with the combination of radiotherapy for the localized and sustained treatment of HCC. The results showed that SOR was released continuously from Gel-SOR-LUF-SeNPs with the degradation of the hydrogel for a prolonged period (over 15 days). The combination of localized and chemoradiotherapy accelerated the apoptosis of HepG2 cells through reducing the expression of Ki67 and CD34, and activating caspase-3 signaling pathway. Further studies demonstrated that this nanosystem showed site-specific and long-term anticancer effects in mice up to 21 days after single subcutaneous injection, and no obvious side effects of mice were found. Taken together, this study presents a local and long-term treatment for HCC, which may shed light on unresectable HCC therapy in the future.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/terapia , Preparaciones de Acción Retardada/química , Neoplasias Hepáticas/terapia , Selenio/administración & dosificación , Sorafenib/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Quimioradioterapia , Femenino , Humanos , Hidrogeles/química , Neoplasias Hepáticas/patología , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/uso terapéutico , Selenio/uso terapéutico , Sorafenib/uso terapéutico
20.
Colloids Surf B Biointerfaces ; 172: 298-307, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30173097

RESUMEN

Functionalized-nanoparticles have been developed as novel therapeutic delivery platform for simultaneous drug loading and therapy over the past decade. Rationally-designed biocompatible nanosystem simultaneously with multistimuli-responsive property and synergistic therapeutic potential are highly desirable for modern biological applications. Herein, Cu2Se nanoparticles (Cu2SeNPs) with suitable size have been functionalized by bull serum albumin (BSA) through a simply, facile and controllable method. As a result, Cu2SeNPs modified by BSA (BSA-Cu2SeNPs) showed excellent biocompatibility and stability. The strong absorbance of BSA-Cu2SeNPs at near infrared region imparts them with high photothermal efficiency. Then loading doxorubicin (DOX, anticancer drug) on the surface of BSA-Cu2SeNPs, and consequently, a novel multifunctional nanosystem of BSA-Cu2SeNPs-DOX is designed. The BSA-Cu2SeNPs can achieve high DOX loading capacity (approximately 157 µg DOX per mg of Cu2Se). Furthermore, a rational and precise release of DOX from the BSA-Cu2SeNPs-DOX could be easily realized under the stimulates of the pH and temperature, which remarkably improved antitumor efficacy of combined chemotherapy and photothermal therapy triggered by 808 nm NIR laser. Thus, the BSA-Cu2SeNPs-DOX could serve as an ideal nanoplatform for cancer diagnosis and treatment in future. The results of cell experiments show that the BSA-Cu2SeNPs-DOX exhibited favorable selective cellular uptake cells. Under the NIR laser irradiation, BSA-Cu2SeNPs-DOX could induce the excessive expression of ROS, eventually leading to the death of U251 cells. Both in vitro and in vivo experiments indicate that the nanosystem of BSA-Cu2SeNPs-DOX showed excellent synergistic therapeutic effect and multistimuli-responsive drug vehicle, which will exert huge potential for future clinical application.


Asunto(s)
Doxorrubicina/farmacología , Portadores de Fármacos/química , Hipertermia Inducida , Nanopartículas/química , Fototerapia , Albúmina Sérica Bovina/química , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Doxorrubicina/química , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Humanos , Nanopartículas/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Espectroscopía Infrarroja por Transformada de Fourier , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA