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












Base de datos
Intervalo de año de publicación
1.
J Mater Chem B ; 12(15): 3741-3750, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38530281

RESUMEN

Oncolytic virus ablation of tumor cells has the advantages of high tumor selectivity, strong immunogenicity, and low side effects. However, the recognition and clearance of oncolytic viruses by the immune system are the main factors limiting their anti-tumor efficiency. As a highly biosafe and highly modifiable oncolytic virus vector, acrylamide can improve the long-term circulation of oncolytic viruses. Still, it is limited in its uptake efficiency by tumor cells. Herein, we constructed an N-hydroxymethyl acrylamide-b-(N-3-aminopropyl methacrylamide)-b-DMC block copolymer (NMA-b-APMA-b-DMA, NAD) as an oncolytic virus carrier, which not only improves the long-term circulation of oncolytic viruses in the body but also shows excellent stability for loading an oncolytic virus. The data shows that there was no obvious difference in the transfection effect of the NAD/Ad complex with or without neutralizing antibodies in the medium, which meant that the cationic carrier mediated by NAD/Ad had good serum stability. Only 10 micrograms of NAD carrier are needed to load the oncolytic virus, which can increase the transfection efficiency by 50 times. Cell experiments and mouse animal experiments show that NAD vectors can significantly enhance the anti-tumor effect of oncolytic viruses. We hope that this work will promote the application of acrylamide as an oncolytic virus vector and provide new ideas for methods to modify acrylamide for biomedical applications.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Ratones , Metionina , Acrilamida , Polímeros , NAD , Acrilamidas , Neoplasias/tratamiento farmacológico , Racemetionina
2.
J Colloid Interface Sci ; 658: 597-609, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38134668

RESUMEN

Non-centrosymmetric tetragonal barium titanate nanocrystals have the potential to serve as piezoelectric catalysts in cancer therapy. When exposed to ultrasound irradiation, BaTiO3 can generate reactive oxygen species with a noninvasive and deep tissue-penetrating approach. However, the application of BaTiO3 in cancer nanomedicine is limited by their biosafety, biocompatibility, and dosage efficiency. To explore the potential application of BaTiO3 in nanomedical cancer treatment, we introduced ultra-small Au nanoparticles onto the surface of BaTiO3 to enhance the piezoelectric catalytic performance. Additionally, we also coated the BaTiO3 with polydopamine to improve their biosafety and biocompatibility. This led to the preparation of a novel multifunctional BaTiO3-based nanoplatform called BTAPs. In vitro and in vivo experiments demonstrated that the incorporation of Au dopants and polydopamine coating successfully improved the piezoelectric catalysis properties and biocompatibility of BaTiO3. Compared with unmodified BaTiO3, BTAPs achieved a similar piezoelectric catalytic effect at a low dose (0.3 mg ml-1 in vitro and 10 mg kg-1 in vivo). Moreover, BTAPs also exhibited enhanced properties in computed tomography imaging and photothermal effects in vivo. Therefore, BTAPs offer valuable insights into the advantages and limitations of piezoelectric catalytic nanomedicine in cancer treatment.


Asunto(s)
Indoles , Nanopartículas del Metal , Neoplasias , Oro/farmacología , Oro/química , Nanopartículas del Metal/química , Polímeros/química , Tomografía Computarizada por Rayos X
3.
J Mater Chem B ; 11(38): 9185-9200, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37724440

RESUMEN

Phototherapy has garnered worldwide attention for its minimal invasiveness, controllability, and spatial selectivity in treating cancer. One promising approach involves the use of near-infrared dye IR780, which demonstrates both photodynamic therapy (PDT) and photothermal therapy (PTT) effects under 808 nm laser irradiation. However, this hydrophobic dye's toxicity and limited tumor targeting ability severely hamper its suitability for cancer applications. Herein, a biocompatible nanoplatform CoOOH-IR780@BSA (CoIRB) is developed to efficiently deliver IR780 and provide multi-mode treatments for colon tumors. Due to the nanocarrier coating, CoIRB nanoparticles demonstrated reliable dispersion and stability, and their biotoxicity was substantially reduced for safer blood circulation, which overcame the biological barrier of IR780. The nanoplatform has also shown considerable results in phototherapy in vivo and in vitro experiments, with successful inhibition of MC38 tumor growth through intravenous administration. Additionally, the introduction of cobalt ions could induce Fenton-like reactions to activate the production of toxic hydroxyl radicals (˙OH), exerting an assisted chemodynamic therapy (CDT) effect. Notably, these nanodrugs also exhibited potential as scavengers of reductive glutathione (GSH) and hydrogen sulfide (H2S), leading to amplifying oxidative damage of reactive oxygen species (ROS). Overall, the versatile therapeutic platform, CoIRB, has opened up considerable prospects as a biotherapeutic option for combining PDT/PTT/CDT against colon cancer.


Asunto(s)
Neoplasias del Colon , Nanosferas , Fotoquimioterapia , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Fotoquimioterapia/métodos , Yoduros , Fototerapia/métodos , Cobalto/farmacología , Neoplasias del Colon/tratamiento farmacológico , Hidróxidos
4.
J Colloid Interface Sci ; 638: 375-391, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36746055

RESUMEN

Lenvatinib (LT), a first-line molecular targeted therapeutic drug for hepatocellular carcinoma (HCC), has been replacing the status of Sorafenib (SF) as the clinically preferred and irreplaceable treatment for a decade. To overcome the low drug utilization and limited single efficacy of LT, ultrasmall copper sulfide nanocrystals (Cu2-xS NCs), and ultrasmall gold nanoparticle (AuNPs) were evenly wrapped into galactosamine conjugated poly(lactide-co-glycolide) (PLGA) as the drug delivery nanoparticles (CAL@PG) by nanoprecipitation. The CAL@PG NPs exhibited excellent stability under physiological conditions, whereas they released LT rapidly in the unique tumor microenvironment (TME) and high temperature, which could be provided by the near-infrared-II (NIR-II) photothermal effect of Cu2-xS NCs. Moreover, the temperature elevation, regenerated hydrogen peroxide (H2O2), and lower pH of TME could substantially boost the reaction potency of copper Fenton-like chemistry. More importantly, this combined therapy significantly improved the efficacy of LT, provided a multifunctional LT delivery system, and enriched the nanoparticle-augmented multimodal synergistic HCC therapy modality.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas del Metal , Nanopartículas , Neoplasias , Humanos , Oro , Nanomedicina Teranóstica , Cobre/química , Peróxido de Hidrógeno , Nanopartículas/química , Línea Celular Tumoral , Fototerapia , Microambiente Tumoral
5.
J Nanobiotechnology ; 20(1): 457, 2022 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-36274142

RESUMEN

Due to the urgent demand for more anti-cancer methods, the new applications of metal ions in cancer have attracted increasing attention. Especially the three kinds of the new mode of cell death, including ferroptosis, calcicoptosis, and cuproptosis, are of great concern. Meanwhile, many metal ions have been found to induce cell death through different approaches, such as interfering with osmotic pressure, triggering biocatalysis, activating immune pathways, and generating the prooxidant effect. Therefore, varieties of new strategies based on the above approaches have been studied and applied for anti-cancer applications. Moreover, many contrast agents based on metal ions have gradually become the core components of the bioimaging technologies, such as MRI, CT, and fluorescence imaging, which exhibit guiding significance for cancer diagnosis. Besides, the new nano-theranostic platforms based on metal ions have experimentally shown efficient response to endogenous and exogenous stimuli, which realizes simultaneous cancer therapy and diagnosis through a more controlled nano-system. However, most metal-based agents have still been in the early stages, and controlled clinical trials are necessary to confirm or not the current expectations. This article will focus on these new explorations based on metal ions, hoping to provide some theoretical support for more anti-cancer ideas.


Asunto(s)
Medios de Contraste , Neoplasias , Humanos , Iones , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Metales/uso terapéutico
6.
J Nanobiotechnology ; 20(1): 415, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109734

RESUMEN

Gastrointestinal cancer (GIC) is a common malignant tumour of the digestive system that seriously threatens human health. Due to the unique organ structure of the gastrointestinal tract, endoscopic and MRI diagnoses of GIC in the clinic share the problem of low sensitivity. The ineffectiveness of drugs and high recurrence rates in surgical and drug therapies are the main factors that impact the curative effect in GIC patients. Therefore, there is an urgent need to improve diagnostic accuracies and treatment efficiencies. Nanotechnology is widely used in the diagnosis and treatment of GIC by virtue of its unique size advantages and extensive modifiability. In the diagnosis and treatment of clinical GIC, surface-enhanced Raman scattering (SERS) nanoparticles, electrochemical nanobiosensors and magnetic nanoparticles, intraoperative imaging nanoparticles, drug delivery systems and other multifunctional nanoparticles have successfully improved the diagnosis and treatment of GIC. It is important to further improve the coordinated development of nanotechnology and GIC diagnosis and treatment. Herein, starting from the clinical diagnosis and treatment of GIC, this review summarizes which nanotechnologies have been applied in clinical diagnosis and treatment of GIC in recent years, and which cannot be applied in clinical practice. We also point out which challenges must be overcome by nanotechnology in the development of the clinical diagnosis and treatment of GIC and discuss how to quickly and safely combine the latest nanotechnology developed in the laboratory with clinical applications. Finally, we hope that this review can provide valuable reference information for researchers who are conducting cross-research on GIC and nanotechnology.


Asunto(s)
Neoplasias Gastrointestinales , Nanopartículas , Sistemas de Liberación de Medicamentos , Detección Precoz del Cáncer , Neoplasias Gastrointestinales/diagnóstico , Neoplasias Gastrointestinales/tratamiento farmacológico , Humanos , Nanopartículas/química , Nanotecnología/métodos
7.
Int J Radiat Oncol Biol Phys ; 114(3): 502-515, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35840114

RESUMEN

PURPOSE: The majority of cancer-related deaths are attributed to metastasis rather than localized primary tumor progression. However, the factors that regulate the premetastatic niche (PMN) and metastasis have not yet been clearly elucidated. We investigated the antimetastatic effects of irradiated tumor cell-derived microparticles (RT-MPs) and highlighted the role of innate immune cells in PMN formation. METHODS AND MATERIALS: Mice were treated 3 times with isolated RT-MPs, followed by tumor cell injection via the tail vein. The hematoxylin and eosin staining was performed to assess the number of tumor nodules in the lungs, and in vivo luciferase-based noninvasive bioluminescence imaging was conducted to detected tumor burden. The mechanisms of RT-MPs mediated PMN formation was evaluated using flow cytometry, transwell assay, and reverse transcription-polymerase chain reaction. RESULTS: RT-MPs inhibited tumor cell colonization in the lungs. Neutrophils phagocytosed RT-MPs and secreted CCL3 and CCL4, which induced monocytes chemotaxis and maturation into macrophages. RT-MPs promoted the transition of neutrophils and macrophages into antitumor phenotypes, hence inhibiting cancer cell colonization and proliferation. CONCLUSIONS: RT-MPs inhibited PMN formation and lung metastasis in a neutrophil- and macrophage-dependent but T cell-independent manner.


Asunto(s)
Micropartículas Derivadas de Células , Neoplasias Pulmonares , Neoplasias Inducidas por Radiación , Animales , Micropartículas Derivadas de Células/patología , Eosina Amarillenta-(YS) , Hematoxilina , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Neoplasias Inducidas por Radiación/patología , Microambiente Tumoral
8.
Comput Intell Neurosci ; 2022: 9046507, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463276

RESUMEN

Pyroptosis is a programmed cell death mediated by gasdermins (GSDMs). The prognostic value of pyroptosis-related genes in different tumor types has been gradually demonstrated recently. However, the prognostic impact of GSDMs expression in glioma remains unclear. Here, we present a comprehensive bioinformatic analysis of gasdermin family member gene expression, producing a prognostic model for glioma and creating a competing endogenous RNA (ceRNA) network. The mRNA expression profiles and clinical information of glioma patients were downloaded from TCGA and CGGA. A risk score based on the gasdermin family was constructed in the TCGA cohort and validated in CGGA. The Jurkat cell was used to verify the relationship between pyroptosis and activation-induced cell death (AICD). We identify a significant association between the expression of GSDMD and GSDME and the glioma stage. The least absolute shrinkage and selection operator (LASSO) Cox regression analysis was used to construct a prognostic gene model based on the four prognostic gasdermin family genes (GSDMC, GSDMD, GSDME, and PJVK). This model was able to predict the overall survival of glioma patients with high accuracy. We show that gasdermin family genes are expressed primarily by immune cells, endothelial cells, and neuronal cells in the tumor microenvironment, rather than by malignant tumor cells. T cells were significantly activated in high-risk patients; however, the activation-induced cell death (AICD) pathway was also significantly activated, suggesting widespread expiration of cytotoxic T lymphocytes (CTLs), facilitating tumor progression. We also identify the lncRNA/miR-296-5p/GSDMD regulatory axis as an important player in glioma progression. We have conducted a comprehensive bioinformatic analysis identifying the importance of gasdermin family members in glioma; a prognostic algorithm containing four genes was constructed.


Asunto(s)
Glioma , MicroARNs , Biomarcadores de Tumor/genética , Biología Computacional , Proteínas de Unión al ADN , Células Endoteliales , Glioma/genética , Humanos , Proteínas Citotóxicas Formadoras de Poros , Piroptosis/genética , Microambiente Tumoral
9.
Nanoscale ; 12(27): 14788-14800, 2020 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-32627781

RESUMEN

Angiogenesis is an irreplaceable therapeutic cancer target, where anti-angiogenesis are drugs that are limited by their hydrophobicity and low therapeutic effects. What is more, the long-term shutdown of tumor blood vessel density also aggravates hypoxia and causes immunosuppression in the tumor microenvironment (TME). In order to solve these shortcomings, we developed a single therapeutic agent based on a bovine serum albumin nanocarrier that can co-deliver the anti-angiogenic drug Sorafenib ("S") and the photosensitizer Ce6 ("C") along with a molecular oxygen supply based on MnO2 ("M") as a convenient one-pot formulated nanoscale agent (SCM@BSA). Compared with anti-angiogenesis monotherapy, SCM@BSA can not only improve upon the solubility and therapeutic effects of anti-angiogenesis agents, but it also reshapes the immunosuppressive TME during anti-angiogenic therapy. Together, these results point out that SCM@BSA synthesized via a very simple method can solve the shortcomings usually experienced during long-term anti-angiogenic therapy.


Asunto(s)
Fotoquimioterapia , Terapia de Inmunosupresión , Compuestos de Manganeso , Óxidos , Oxígeno
10.
J Adv Res ; 24: 353-361, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32489680

RESUMEN

Inorganic or inorganic-organic hybrid nanomaterials have great potential for applications in the biomedical fields. Biological half-life is an essential pharmacokinetic parameter for these materials to function in vivo. Compared to inductively coupled plasma mass spectrometry (ICP-MS), which is the gold standard, laser-induced breakdown spectroscopy (LIBS) is a faster and more efficient elemental detection method. We investigated an efficient way to quantify the metabolic rate using LIBS. Nanoparticle platforms, such as manganese dioxide-bovine serum albumin (MnO2-BSA) or boehmite-bovine serum albumin (AlO(OH)-BSA) were injected into mice through intravenous administration for LIBS spectrum acquisition. First, the spectral background was corrected using the polynomial fitting method; The spectral interference was eliminated by Lorentz fitting for each LIBS spectrum simultaneously. The support vector regression (SVR) was then used for LIBS quantitative analyses. Finally, the LIBS results were compared with the ICP-MS ones. The half-lives of MnO2-BSA calculated by LIBS and ICP-MS were 2.49 and 2.42 h, respectively. For AlO(OH)-BSA, the half-lives detected by LIBS and ICP-MS were 3.46 and 3.57 h, respectively. The relative error of LIBS is within 5% compared to ICP-MS. The results demonstrate that LIBS is a valuable tool for quantifying the metabolic rates with a high degree of accuracy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...