Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 206, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658950

RESUMEN

The insufficient abundance and weak activity of tumour-infiltrating lymphocytes (TILs) are two important reasons for the poor efficacy of PD-1 inhibitors in hepatocellular carcinoma (HCC) treatment. The combined administration of tanshinone IIA (TSA) and astragaloside IV (As) can up-regulate the abundance and activity of TILs by normalising tumour blood vessels and reducing the levels of immunosuppressive factors respectively. For enhancing the efficacy of PD-1 antibody, a magnetic metal-organic framework (MOF) with a homologous tumour cell membrane (Hm) coating (Hm@TSA/As-MOF) is established to co-deliver TSA&As into the HCC microenvironment. Hm@TSA/As-MOF is a spherical nanoparticle and has a high total drug-loading capacity of 16.13 wt%. The Hm coating and magnetic responsiveness of Hm@TSA/As-MOF provide a homologous-magnetic dual-targeting, which enable Hm@TSA/As-MOF to counteract the interference posed by ascites tumour cells and enhance the precision of targeting solid tumours. Hm coating also enable Hm@TSA/As-MOF to evade immune clearance by macrophages. The release of TSA&As from Hm@TSA/As-MOF can be accelerated by HCC microenvironment, thereby up-regulating the abundance and activity of TILs to synergistic PD-1 antibody against HCC. This study presents a nanoplatform to improve the efficacy of PD-1 inhibitors in HCC, providing a novel approach for anti-tumour immunotherapy in clinical practice.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Estructuras Metalorgánicas , Receptor de Muerte Celular Programada 1 , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Carcinoma Hepatocelular/tratamiento farmacológico , Animales , Ratones , Humanos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Línea Celular Tumoral , Inhibidores de Puntos de Control Inmunológico/farmacología , Microambiente Tumoral/efectos de los fármacos , Ratones Endogámicos BALB C , Saponinas/farmacología , Saponinas/química , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología
2.
Adv Healthc Mater ; 13(14): e2303659, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38386849

RESUMEN

Sustainable retinal codelivery poses significant challenges technically, although it is imperative for synergistic treatment of wet age-related macular degeneration (wAMD). Here, a microemulsion-doped hydrogel (Bor/PT-M@TRG) is engineered as an intravitreal depot composing of temperature-responsive hydrogel (TRG) and borneol-decorated paeoniflorin (PF) & tetramethylpyrazine (TMP)-coloaded microemulsions (Bor/PT-M). Bor/PT-M@TRG, functioning as the "ammunition depot", resides in the vitreous and continuously releases Bor/PT-M as the therapeutic "bullet", enabling deep penetration into the retina for 21 days. A single intravitreal injection of Bor/PT-M@TRG yields substantial reductions in choroidal neovascularization (CNV, a hallmark feature of wAMD) progression and mitigates oxidative stress-induced damage in vivo. Combinational PF&TMP regulates the "reactive oxygen species/nuclear factor erythroid-2-related factor 2/heme oxygenase-1" pathway and blocks the "hypoxia inducible factor-1α/vascular endothelial growth factor" signaling in retina, synergistically cutting off the loop of CNV formation. Utilizing fluorescence resonance energy transfer and liquid chromatography-mass spectrometry techniques, they present compelling multifaceted evidence of sustainable retinal codelivery spanning formulations, ARPE-19 cells, in vivo eye balls, and ex vivo section/retina-choroid complex cell levels. Such codelivery approach is elucidated as the key driving force behind the exceptional therapeutic outcomes of Bor/PT-M@TRG. These findings highlight the significance of sustainable retinal drug codelivery and rational combination for effective treatment of wAMD.


Asunto(s)
Pirazinas , Animales , Pirazinas/química , Pirazinas/administración & dosificación , Pirazinas/farmacología , Pirazinas/farmacocinética , Retina/efectos de los fármacos , Retina/metabolismo , Degeneración Macular/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Humanos , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Ratones , Hidrogeles/química , Hidrogeles/farmacología , Estrés Oxidativo/efectos de los fármacos , Canfanos/química , Canfanos/farmacología , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
3.
J Ethnopharmacol ; 325: 117869, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38342153

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Coronary heart disease (CHD) is a chronic disease that seriously threatens people's health and even their lives. Currently, there is no ideal drug without side effects for the treatment of CHD. Trichosanthis Pericarpium (TP) has been used for several years in the treatment of diseases associated with CHD. However, there is still a need for systematic research to unravel the pharmacodynamic substances and possible mechanism of TP in the treatment of coronary heart. AIM OF THE STUDY: The purpose of current study was to explore the pharmacodynamic substances and potential mechanisms of TP in the treatment of CHD via integrating network pharmacology with plasma pharmacochemistry and experimental validation. MATERIALS AND METHODS: The effect of TP intervention in CHD was firstly assessed on high-fat diet combined with isoprenaline-induced CHD rats and H2O2-induced H9c2 cells, respectively. Then, the LC-MS was utilized to identify the absorbed components of TP in the plasma of CHD rats, and this was used to develop a network pharmacology prediction to obtain the possible active components and mechanisms of action. Molecular docking and immunohistochemistry were used to explore the interaction between TP and key targets. Subsequently, the efficacy of the active ingredients was investigated by in vitro cellular experiments, and their metabolic pathways in CHD rats were further analyzed. RESULTS: The effects of TP on amelioration of CHD were verified by in vivo and in vitro experiments. Plasma pharmacochemistry and network pharmacology screened six active components in plasma including apigenin, phenylalanine, quercetin, linoleic acid, luteolin, and tangeretin. The interaction of these compounds with potential key targets AKT1, IL-1ß, IL-6, TNF-α and VEGFA were preliminarily verified by molecular docking. And immunohistochemical results showed that TP reduced the expression of AKT1, IL-1ß, IL-6, TNF-α and VEGFA in CHD rat hearts. Then cellular experiments confirmed that apigenin, phenylalanine, quercetin, linoleic acid, luteolin, and tangeretin were able to reduce the ROS level in H2O2-induced HUVEC cells and promote the migration and tubule formation of HUVEC cells, indicating the pharmacodynamic effects of the active components. Meanwhile, the metabolites of TP in CHD rats suggested that the pharmacological effects of TP might be the result of the combined effects of the active ingredients and their metabolites. CONCLUSION: Our study found that TP intervention in CHD is characterized by multi-component and multi-target regulation. Apigenin, phenylalanine, linoleic acid, quercetin, luteolin, and tangeretin are the main active components of TP. TP could reduce inflammatory response and endothelial damage by regulating AKT1, IL-1ß, IL-6, TNF-α and VEGFA, reduce ROS level to alleviate the oxidative stress situation and improve heart disease by promoting angiogenesis to regulate endothelial function. This study also provides an experimental and scientific basis for the clinical application and rational development of TP.


Asunto(s)
Enfermedad Coronaria , Medicamentos Herbarios Chinos , Humanos , Animales , Ratas , Apigenina , Luteolina/farmacología , Luteolina/uso terapéutico , Peróxido de Hidrógeno , Interleucina-6 , Ácido Linoleico , Simulación del Acoplamiento Molecular , Farmacología en Red , Quercetina , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa , Enfermedad Coronaria/tratamiento farmacológico , Interleucina-1beta , Fenilalanina
4.
ACS Nano ; 17(23): 23829-23849, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37991391

RESUMEN

Programmed cell death protein 1 (PD-1) inhibitors are the most common immune-checkpoint inhibitors and considered promising drugs for hepatocellular carcinoma (HCC). However, in clinical settings, they have a low objective response rate (15%-20%) for patients with HCC; this is because of the insufficient level and activity of tumor-infiltrating T lymphocytes (TILs). The combined administration of oxymatrine (Om) and astragaloside IV (As) can increase the levels of TILs by inhibiting the activation of cancer-associated fibroblasts (CAFs) and improve the activity of TILs by enhancing their mitochondrial function. In the present study, we constructed a magnetic metal-organic framework (MOF)-based nanoplatform with platelet membrane (Pm) coating (PmMN@Om&As) to simultaneously deliver Om and As into the HCC microenvironment. We observed that PmMN@Om&As exhibited a high total drug-loading capacity (33.77 wt %) and good immune escape. Furthermore, it can target HCC tissues in a magnetic field and exert long-lasting effects. The HCC microenvironment accelerated the disintegration of PmMN@Om&As and the release of Om&As, thereby increasing the level and activity of TILs by regulating CAFs and the mitochondrial function of TILs. In addition, the carrier could synergize with Om&As by enhancing the oxygen consumption rate and proton efflux rate of TILs, thereby upregulating the mitochondrial function of TILs. Combination therapy with PmMN@Om&As and α-PD-1 resulted in a tumor suppression rate of 84.15% and prolonged the survival time of mice. Our study provides a promising approach to improving the antitumor effect of immunotherapy in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Estructuras Metalorgánicas , Humanos , Animales , Ratones , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Receptor de Muerte Celular Programada 1 , Estructuras Metalorgánicas/farmacología , Fenómenos Magnéticos , Microambiente Tumoral , Linfocitos T CD8-positivos
5.
Int J Biol Macromol ; 251: 126323, 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37586629

RESUMEN

M1 polarization of tumor-associated macrophages (TAMs) is a promising approach to breaking through therapeutic barriers imposed by the immunosuppressive tumor microenvironment (TME). As a clinically-used immunopotentiator for cancer patients after chemotherapies; however, the immunomodulatory mechanism and potential of polyporus polysaccharide (PPS) remains unclear. Here, we present mannose-decorated PPS-loaded superparamagnetic iron-based nanocomposites (Man/PPS-SPIONs) for synergistic M1 polarization of TAMs and consequent combinational anti-breast cancer therapy. Once internalized by M2-like TAMs, PPS released from Man/PPS-SPIONs induces the M1 polarization via IFN-γ secretion and downstream NF-κB pathway activating. The SPIONs within the nanocomposites mediate a Fenton reaction, producing OH· and activating the subsequent NF-κB/MAPK pathway, further facilitating the M1 polarization. The Man/PPS-SPIONs thereby establish a positive feedback loop of M1 polarization driven by the "IFN-γ-Fenton-NF-κB/MAPK" multi-pathway, leading to a series of anti-tumoral immunologic responses in the TME and holding promising potential in combinational anticancer therapies. Our study offers a new strategy to amplify TME engineering by combinational natural carbohydrate polymers and iron-based materials.

6.
Int J Nanomedicine ; 18: 2839-2853, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37273286

RESUMEN

Background: Berberine (BR) shows promise as a candidate for treating irritable bowel syndrome with diarrhea (IBS-D). However, the undesired physicochemical properties and poor oral absorption limit its clinical translation. A ketogenic diet (KD) can induce intestinal overexpression of cannabidiol (CB) receptors, which may offer a potential target for IBS-D-specific delivery of BR. Methods: The microemulsions loaded with BR and decorated with cannabidiol (CBD/BR-MEs) were developed through a one-step emulsion method. The pharmaceutical behaviors of the CBD/BR-MEs were measured using dynamic light scattering and high-performance liquid chromatography. The efficacy of the anti-IBS-D therapy was evaluated by assessing fecal water content, Bristol score, and AWR score. The intestinal permeability were assessed through immunofluorescent staining of CB1 and ZO-1, respectively. The signaling of CREB/BDNF/c-Fos was also studied along with immunofluorescent and immunohistochemical examination of brain sections. Results: The CBD/BR-MEs, which had a particle size of approximately 30 nm and a surface density of 2% (wt%) CBD, achieved greater than 80% (wt%) encapsulation efficiency of BR. The pharmacokinetics performance of CBD/BR-MEs was significantly improved in the KD-fed IBS-D rats than the standard diet-fed ones, which is highly related to intestinal expression of CB1 receptors. The treatment with CBD/BR-MEs and KD exhibited evident comprehensive advantages over the other groups in terms of anti-IBS-D efficacy. CBD/BR-MEs and KD synergistically decreased intestinal permeability. Moreover, the treatment with CBD/BR-MEs and KD not only blocked the CREB/BDNF/c-Fos signaling in the brain but also decreased the levels of neurotrophic factors, neurotransmitters, and inflammatory cytokines in the serum of IBS-D model rats. Conclusion: Such a design represents the first attempt at IBS-D-targeted drug delivery for improved oral absorption and efficacy through KD-induced target exposure, which holds promising potential for the treatment of IBS-D.


Asunto(s)
Berberina , Cannabidiol , Dieta Cetogénica , Síndrome del Colon Irritable , Ratas , Animales , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/metabolismo , Berberina/farmacología , Berberina/uso terapéutico , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo , Diarrea/tratamiento farmacológico
7.
Biomater Res ; 27(1): 48, 2023 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-37198657

RESUMEN

BACKGROUND: Conventional dissolving microneedles (DMNs) face significant challenges in anti-melanoma therapy due to the lack of active thrust to achieve efficient transdermal drug delivery and intra-tumoral penetration. METHODS: In this study, the effervescent cannabidiol solid dispersion-doped dissolving microneedles (Ef/CBD-SD@DMNs) composed of the combined effervescent components (CaCO3 & NaHCO3) and CBD-based solid dispersion (CBD-SD) were facilely fabricated by the "one-step micro-molding" method for boosted transdermal and tumoral delivery of cannabidiol (CBD). RESULTS: Upon pressing into the skin, Ef/CBD-SD@DMNs rapidly produce CO2 bubbles through proton elimination, significantly enhancing the skin permeation and tumoral penetration of CBD. Once reaching the tumors, Ef/CBD-SD@DMNs can activate transient receptor potential vanilloid 1 (TRPV1) to increase Ca2+ influx and inhibit the downstream NFATc1-ATF3 signal to induce cell apoptosis. Additionally, Ef/CBD-SD@DMNs raise intra-tumoral pH environment to trigger the engineering of the tumor microenvironment (TME), including the M1 polarization of tumor-associated macrophages (TAMs) and increase of T cells infiltration. The introduction of Ca2+ can not only amplify the effervescent effect but also provide sufficient Ca2+ with CBD to potentiate the anti-melanoma efficacy. Such a "one stone, two birds" strategy combines the advantages of effervescent effects on transdermal delivery and TME regulation, creating favorable therapeutic conditions for CBD to obtain stronger inhibition of melanoma growth in vitro and in vivo. CONCLUSIONS: This study holds promising potential in the transdermal delivery of CBD for melanoma therapy and offers a facile tool for transdermal therapies of skin tumors.

8.
J Nanobiotechnology ; 21(1): 71, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36859261

RESUMEN

Sustained retina drug delivery and rational drug combination are considered essential for enhancing the efficacy of therapy for wet age-related macular degeneration (wAMD) due to the conservative structure of the posterior ocular segment and the multi-factorial pathological mechanism. Designing a drug co-delivery system that can simultaneously achieve deep penetration and long-lasting retention in the vitreous is highly desired, yet remains a huge challenge. In this study, we fabricated Bor/RB-M@TRG as an intravitreal-injectable hydrogel depot for deep penetration into the posterior ocular segment and long-lasting distribution in the retinal pigment epithelium (RPE) layer. The Bor/RB-M@TRG consisted of borneol-decorated rhein and baicalein-coloaded microemulsions (Bor/RB-M, the therapy entity) and a temperature-responsive hydrogel matrix (the intravitreal depot). Bor/RB-M exhibited the strongest in vitro anti-angiogenic effects among all the groups studied, which is potentially associated with improved cellular uptake, as well as the synergism of rhein and baicalein, acting via anti-angiogenic and anti-oxidative stress pathways, respectively. Importantly, a single intravitreal (IVT) injection with Bor/RB-M@TRG displayed significant inhibition against the CNV of wAMD model mice, compared to all other groups. Particularly, coumarin-6-labeled Bor/RB-M@TRG (Bor/C6-M@TRG) could not only deeply penetrate into the retina but also stably accumulate in the RPE layer for at least 14 days. Our design integrates the advantages of borneol-decorated microemulsions and hydrogel depots, offering a promising new approach for clinically-translatable retinal drug delivery and synergistic anti-wAMD treatment.


Asunto(s)
Hidrogeles , Retina , Animales , Ratones , Antraquinonas
9.
Drug Deliv ; 29(1): 3454-3466, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36447364

RESUMEN

Combinational icaritin (IC) and coix seed oil (CSO) holds promising potential in the treatment of hepatocellular carcinoma. However, traditional cocktail therapy is facing difficulties to optimize the synergistic antitumor efficacy due to the asynchronous pharmacokinetics. Therefore, we developed an icaritin-loaded microemulsion based on coix seed oil (IC-MEs) for improved pharmacokinetics and enhanced antitumor efficacy. The preparation technology of IC-MEs was optimized by the Box-Behnken design and the pharmaceutical properties were characterized in detail. IC-MEs show synergistic antiproliferation against HepG2 cells compared with monotherapy. The mechanism is associated with stronger apoptosis induction via enhancing caspases-3 activity. IC-MEs significantly improve the bioavailability of IC due to the encapsulation of coix oil-based microemulsion and also obtain the desired liver accumulation and elimination. More importantly, IC-MEs exhibit the overwhelming antitumor ability among all of the treatments on the HepG2 xenograft-bearing mice. This study verifies the feasibility of using coix oil-based microemulsion to improve the antitumor effect of water-insoluble components.


Asunto(s)
Coix , Neoplasias Hepáticas , Humanos , Animales , Ratones , Flavonoides/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Aceites de Plantas/farmacología
10.
J Nanobiotechnology ; 20(1): 114, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35248071

RESUMEN

Cancer-associated fibroblasts (CAFs) deteriorate tumor microenvironment (TME) and hinder intra-tumoral drug delivery. Direct depleting CAFs exists unpredictable risks of tumor metastasis. Epithelial-mesenchymal transition (EMT) is a critical process of CAFs converted from hepatic stellate cells during hepatocellular tumorigenesis; however, until now the feasibility of reversing EMT to battle hepatocellular carcinoma has not been comprehensively explored. In this study, we report a CFH peptide (CFHKHKSPALSPVGGG)-decorated liposomal oxymatrine (CFH/OM-L) with a high affinity to Tenascin-C for targeted inactivating CAFs through reversing EMT, which is verified by the upregulation of E-cadherin and downregulation of vimentin, N-cadherin, and snail protein in vivo and in vitro. After the combination with icaritin-loaded lipid complex, CFH/OM-L obviously boosts the comprehensive anticancer efficacy in both 3D tumor spheroids and stromal-rich tumor xenograft nude mouse models. The combinational therapy not only effectively reversed the in vivo EMT process but also significantly lowered the collagen, creating favorable conditions for deep penetration of nanoparticles. More importantly, CFH/OM-L does not kill but inactivates CAFs, resulting in not only a low risk of tumor metastasis but also a reprogramming TME, such as M1 tumor-associated macrophages polarization and natural killer cells activation. Such strategy paves a moderate way to remold TME without depleting CAFs and provides a powerful tool to design strategies of combinational hepatocellular carcinoma therapy.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Hepatocelular , Neoplasias Hepáticas , Alcaloides , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/fisiología , Fibroblastos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Péptidos/metabolismo , Péptidos/farmacología , Quinolizinas , Microambiente Tumoral
11.
J Drug Target ; 30(6): 646-656, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35225125

RESUMEN

Myocardial ischaemia-reperfusion injury (MI/RI) induces injury against cardiomyocytes and triggers myocardial infarction. Previously, we demonstrated that tetramethylprazine (TMP) was a promising therapeutic agent for attenuating MI/RI. However, poor absorption and low homing efficiency are two main obstacles to the further application of TMP. In this study, a platelet membrane-cloaking TMP-loaded microemulsion (P/TMP-MEs) capable of promoting in vivo absorption and decreasing non-targeted accumulation was fabricated for the improved MI/RI therapy. The average particle size and zeta potential of P/TMP-MEs were 35.9 ± 2.5 nm and -29.4 ± 3.1 mV, respectively. 35.4 ± 2.4 wt% TMP was released from P/TMP-MEs after 48 h of incubation with rat plasma. The coating of the platelet membrane significantly decreased the internalisation of P/TMP-MEs by THP-1 macrophage-like cells compared with the non-platelet modified TMP-loaded microemulsion (TMP-MEs). Besides, P/TMP-MEs did not activate the complement system. After treatment with P/TMP-MEs, the dehydrogenase (LDH) level of the cardiomyocytes was significantly lower than other controls. Rats single-administrated with P/TMP-MEs exhibited the area under the plasma concentration-time curve (AUC) at 463796.7 ± 53614.3 ng/mL/h, and ∼400 ng/mL TMP could still be detected from the plasma after 24 h of administration, exhibiting a prolonged blood circulation time as the platelet membrane coating. More importantly, in the MI/RI therapy in vivo, the creatine kinase (CK) and LDH of the rats treated with P/TMP-MEs were remarkably decreased compared with the free TMP and TMP-MEs groups. The combinational strategy of platelet membrane coating and microemulsion assembly endows TMP with a better prospect for MI/RI therapy.


Asunto(s)
Infarto del Miocardio , Daño por Reperfusión Miocárdica , Animales , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley
12.
Int J Pharm ; 605: 120798, 2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34126177

RESUMEN

Amphiphilic chitosan derivatives have attracted wide attention as drug carriers due to their physicochemical properties. However, obtaining a desired amphiphilic chitosan derivative by tuning the various functional groups was complex and time-consuming. Therefore, a facile and common synthesis strategy would be promising. In this study, a modular strategy based on strain-promoted azide-alkyne cycloaddition (SPAAC) click reaction was designed and applied in synthesizing deoxycholic acid- or octanoic acid-modified N-azido propionyl-N,O-sulfate chitosan through tuning the hydrophobic groups. Additionally, chitosan derivatives with the same substitute groups were prepared via amide coupling as controls. We demonstrated that these derivates via the two strategies showed no obvious difference in physicochemical properties, drug loading ability and biosafety, indicating the feasibility of modular strategy. Notably, the modular strategy exhibited advantages including high reactivity, flexibility and reproducibility. We believe that this modular strategy could provide varied chitosan derivatives in an easy and high-efficiency way for improving multifunctional drug carriers.


Asunto(s)
Quitosano , Azidas , Química Clic , Portadores de Fármacos , Reproducibilidad de los Resultados
13.
Mol Pharm ; 18(3): 822-835, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33586986

RESUMEN

Lung metastasis is a fatal and late-stage event for many solid tumors. Multiple lines of evidence have demonstrated that diallyl trisulfide (DATS), an active ingredient of garlic, possesses striking antimetastatic effects. However, the lack of highly efficient organ-compatible carriers restricts its application. In the present study, we showed that extracellular microparticles encapsulated with DATS (DATS-MPs) were capable of interfering with the prometastatic inflammatory microenvironment in local tissues. DATS-MPs were successfully prepared and exhibited typical characteristics of B16BL6-derived extracellular vesicles. The DATS-MPs preferentially fused with cancer cells and endogenous cells (mouse lung epithelial MLE-12 cells) from the metastatic organs in vitro. More interestingly, the systemically administered MPs predominantly accumulated in the lung tissue that serves as their main metastatic organ. The drug-loaded MPs exerted higher antimetastatic effects than DATS alone in both the spontaneous and the experimental metastasis models in mice (*p < 0.05). Additionally, we found that DATS-MPs inhibited tumor cell migration and interfered with the prometastatic inflammatory microenvironment via decreasing the release of S100A8/A9, serum amyloid A (SAA), and interleukin-6 (IL-6) and inhibiting the expression of fibronectin, MRP8, myeloperoxidase (MPO), and the toll-like receptor 4 (TLR4)-Myd88 in the lung tissues. Collectively, DATS-MPs appeared to enhance the antimetastatic efficiency of DATS in animal models under study.


Asunto(s)
Compuestos Alílicos/farmacología , Vesículas Extracelulares/metabolismo , Inflamación/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Sulfuros/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Calgranulina A/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibronectinas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Melanoma/metabolismo , Melanoma/patología , Ratones , Peroxidasa/metabolismo , Receptor Toll-Like 4/metabolismo
14.
Drug Deliv ; 27(1): 1608-1624, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33179521

RESUMEN

The accumulation and penetration of antitumor drugs in tumor tissues are directly related to their antitumor effects. The particle size of the nanodrug delivery system is one of the most important factors for the accumulation and penetration of antitumor drugs within tumor tissues. Generally, nanodelivery systems of intermediate size (100-120 nm) are capable of efficient accumulation owing to prolonged circulation and enhanced permeability and retention (EPR) effect; however, smaller ones (20-40 nm) are effective for deep penetration within tumor tissue. Currently a conventional drug delivery system cannot possess two types of optimal sizes, simultaneously. To solve this and to enhance cervical cancer treatment, a furin-responsive triterpenine-based liposomal complex (PEGcleavable Tf-CTM/L), with Tf-CTM (transferrin-modified tripterine-loaded coix seed oil microemulsion) in core, coated with a thermo-sensitive lipid and a kind of PEG shell modified with a furin-cleavable peptide was developed to improve tumor-specific accumulation and penetration. Herein, PEGcleavable Tf-CTM/L was capable of efficient accumulation because of EPR effect. The PEG shells could timely detach under stimulation of overexpressed furin protein to solve the problem of the steric hindrance dilemma. The small-sized Tf-CTM released under stimulation of tumor microthermal environment in cervical cancer, which was efficient with regards to deep penetration at tumor sites. Notably, compared to the use of triterpenine alone, PEGcleavable Tf-CTM/L promoted anticervical efficacy and displayed diminished systemic toxicity by efficient accumulation and deep penetration of antitumor drugs within tumor tissues. Our study provides a new strategy, and holds promising potential for anticervical cancer treatment.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Furina/metabolismo , Liposomas/química , Triterpenos Pentacíclicos/química , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Coix/química , Sistemas de Liberación de Medicamentos/métodos , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Tamaño de la Partícula , Transferrina/metabolismo
15.
Biomater Sci ; 8(14): 3916-3925, 2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32555847

RESUMEN

Increasing evidence indicates that the tumor microenvironment (TME) imposes various obstacles in response to chemotherapies. Sodium tanshinone IIA sulfonate (STS) has a validated ability to repair the unfavorable TME, providing a suitable environment for celastrol-based chemotherapy. However, remodeling TME still possesses enormous challenges for STS due to the difficulty in a controlled release at tumor sites. Gold nanorods (GNRs) capable of converting near-infrared (NIR) light into heat offer a promising trigger approach to regulate the local drug release. Here, we fabricated a gold nanorod-anchored thermosensitive liposomal complex co-loaded with STS and celastrol (G-T/C-L), which could sequentially release STS and celastrol upon NIR irradiation at 808 nm. When G-T/C-L reaches the sites, NIR illumination produces mild heat (∼43 °C) and thereby triggers a rapid release of STS in the initial stage, decreasing the level of tumoral blood vessels, collagen, cancer-associated fibroblasts, and Th2 type cytokines. In the subsequent stage, celastrol was unloaded to exert an anticancer effect under an activated TME. In proof-of-concept studies, the treatment of G-T/C-L with NIR illumination showed a significant improvement in anticancer efficacy both in vitro and in vivo but without conventional photothermal therapy-associated side effects. This study proposes photothermal-triggered technology to realize controlled drug release, enriching the application with combinational STS and celastrol in anti-breast cancer therapy.


Asunto(s)
Nanotubos , Neoplasias , Línea Celular Tumoral , Doxorrubicina , Oro , Calor , Humanos , Rayos Infrarrojos , Liposomas , Microambiente Tumoral
16.
ACS Appl Mater Interfaces ; 12(29): 32301-32311, 2020 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-32575984

RESUMEN

A fever-mimic response capable of recruiting reprogrammed macrophages holds great potential in the engineering of the tumor microenvironment (TME). Low-temperature photothermal therapy (LT-PTT) can maintain tumors at a fever-like temperature (<45 °C) temporarily; however, it still faces several challenges in efficient regulation of TME because of reciprocal cross-talk between the bypass pathways. Here, we report a synergistic engineering of TME through an enhanced activation of a fever-mimic response based on both LT-PTT and tumor vascular normalization. Such engineering is achieved by a fever-inducible lipid nanocomposite (GNR-T/CM-L), which produces mild heat (∼43 °C) and sequentially releases multicomponents to cooperatively upregulate interferon-gamma under NIR irradiation, forming a bidirectionally closed loop for downstream M1 tumor-associated macrophage polarization and promoting the inhibition of the tumor growth. In proof-of-concept studies, GNR-T/CM-L demonstrated efficient tumor ablation in breast tumor xenograft-bearing mice and significantly prolonged their survival period. It paves an avenue to precisely reprogram TME for efficient cancer therapy through synergistic pathways of creating fever-like responses in the tumor.


Asunto(s)
Antineoplásicos/farmacología , Hipertermia Inducida , Lípidos/farmacología , Nanocompuestos/química , Fármacos Fotosensibilizantes/farmacología , Temperatura , Animales , Antineoplásicos/química , Femenino , Humanos , Rayos Infrarrojos , Lípidos/química , Células MCF-7 , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Desnudos , Imagen Óptica , Tamaño de la Partícula , Fármacos Fotosensibilizantes/química , Terapia Fototérmica , Propiedades de Superficie , Microambiente Tumoral/efectos de los fármacos
17.
Carbohydr Polym ; 229: 115498, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31826492

RESUMEN

Controlled release and tumor-selective distribution are highly desirable for anticancer nanomedicines. Here, we design and synthesize an anisamide-conjugated N-octyl-N,O-maleoyl-O-phosphoryl chitosan (a-OMPC) which can form amphiphilic micelles featuring pH-responsive release and high affinity to sigma-1 receptor-overexpressed tumors for paclitaxel (PTX) delivery. Thereinto, maleoyl and phosphoryl groups cooperatively contribute to pH-responsive drug release due to a conversion from hydrophile to hydrophobe in the acidic microenvironment of endo/lysosomes. We demonstrated that PTX-loaded a-OMPC micelles (PTX-aM) enhanced the cellular internalization via the affinity between anisamide and sigma-1 receptor, rapidly released drug in endo/lysosomes and elevated the cytotoxicity against PC-3 cells. The in vivo studies further verified that PTX-aM could largely accumulate at the tumor site even after 24 h of administration, resulting in obvious inhibition effect and prolonged survival period in PC-3 tumor xenograft-bearing mice. Moreover, OMPC showed no obvious hemolytic and acute toxicity. Collectively, this chitosan derivate holds a promising potential in application of prostate cancer-targeted drug delivery system.


Asunto(s)
Quitosano/química , Interacciones Hidrofóbicas e Hidrofílicas , Terapia Molecular Dirigida , Paclitaxel/química , Paclitaxel/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Receptores sigma/metabolismo , Animales , Quitosano/toxicidad , Preparaciones de Acción Retardada , Portadores de Fármacos/química , Portadores de Fármacos/toxicidad , Regulación Neoplásica de la Expresión Génica , Hemólisis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ensayo de Materiales , Ratones , Micelas , Células PC-3 , Paclitaxel/uso terapéutico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor Sigma-1
18.
Mol Pharm ; 16(12): 4826-4835, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31663764

RESUMEN

Tumor-targeted ligand modification and nanosized coloaded drug delivery systems are promising for cancer therapy. In this study, we showed that coix seed oil and tripterine coloaded microemulsions with a transferrin modification (Tf-CT-MEs) could improve the treatment of cervical cancer. Tf-CT-MEs exhibited good stability in serum and a notably synergistic antiproliferation effect. In the HeLa xenograft tumor-bearing mouse model, Tf-CT-MEs accumulated at tumor sites and penetrated deeply in tumor tissues. Tf-CT-MEs had superior anticancer efficacy in vivo, which greatly slowed the growth of tumors (***p < 0.001 vs saline). We also found that Tf-CT-MEs inhibited tumor cell proliferation, enhanced antiangiogenesis, and induced apoptosis by regulating bax/bcl-2 and the activating caspase-3 pathway. Tf-CT-MEs decreased by 27.7, 26.9, 61.2, and 42.5% of concentrations of TGF-ß1, CCL2, TNF-α, and IL-6 in serum, respectively. In addition, Tf-CT-MEs showed little toxicity in vital organs. These results were due to the improved drug delivery efficiency. Collectively, Tf-CT-MEs enhance tumor-targeting, facilitate deep penetration of drugs, and have promising potential as an efficient treatment for cervical cancer.


Asunto(s)
Coix/química , Emulsiones/farmacología , Aceites de Plantas/farmacología , Semillas/química , Transferrina/farmacología , Triterpenos/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Triterpenos Pentacíclicos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
19.
Am J Transl Res ; 11(9): 5689-5702, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31632540

RESUMEN

Diabetic nephropathy (DN) is known as a major microvascular complication leading cause of end-stage renal disease, it generally followed by the process of podocyte fragmentation and detachment. Transforming growth factor ß1 (TGF-ß1) signaling pathway plays a pivotal role in the initiation and progression of DN. In present study, we aim to investigate the effect of lycopus extracts on podocytes injury and TGF-ß signaling. In present study, lycopus extracts treatment abolished the gain in blood glucose and body weight in a dose dependent manner and possessed protective effect on the renal damage, which was indicated by the decreased concentration of Scr, BUN and urine creatinine of serum. Histopathological examination also demonstrated lycopus extracts exert protective effect on renal damage. Western blotting and immunohistochemical results revealed lycopus extracts treatment upregulated the expression of nephrin and down-regulated the expression levels of TGF-ß1 and Smad4. Moreover, lycopus extracts treatment suppressed TGF-ß1-induced phosphorylation of Smad2/3, ERK1/2 and p38 both in vivo and vitro. In conclusion, lycopus extracts is a novel agent that ameliorate podocytes injury by inhibiting TGF-ß signaling pathway and possess potential therapeutic effect on renal damage of DN rats.

20.
Carbohydr Polym ; 207: 428-439, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30600025

RESUMEN

Herein, we describe a novel amphipathic chitosan derivative (N-octyl-N'-phthalyl-O-phosphoryl chitosan, abbreviated as OPPC) as an effective oral delivery platform for P-gp substrates, especially paclitaxel (PTX). OPPC could readily self-assemble into micelles, solubilize and encapsulate PTX into the hydrophobic inner core of OPPC with superior loading capacity to chitosan. PTX/OPPC micelles possessed improved intestinal epithelial permeability and oral bioavailability of PTX evaluated by in situ perfusion and pharmacokinetic studies. In vivo fluorescence imaging revealed enhanced stability and integrity of OPPC micelles in mice gastrointestine. Furthermore, cellular uptake studies revealed effective transport and accumulation of OPPC micelles loading PTX or rhodamine-123 into Caco-2 cells via clathrin/cavelin-mediated endocytosis and OPPC-mediated P-gp inhibition. Mechanistically, the inhibition of P-gp efflux pumps by OPPC resulted from the reduction of membrane fluidity and decreased P-gp ATPase activity. In summary, OPPC micelles may serve as an efficient and promising delivery system for enhancing oral bioavailability of P-gp substrates.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quitosano/análogos & derivados , Quitosano/química , Portadores de Fármacos/química , Paclitaxel/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Administración Oral , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Células CACO-2 , Quitosano/síntesis química , Quitosano/toxicidad , Regulación hacia Abajo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Masculino , Fluidez de la Membrana/efectos de los fármacos , Ratones Endogámicos BALB C , Micelas , Paclitaxel/administración & dosificación , Paclitaxel/química , Ratas Sprague-Dawley , Solubilidad , Transcitosis/efectos de los fármacos , Verapamilo/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA