Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biomater Sci ; 11(21): 7114-7123, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37671612

RESUMEN

The design of advanced carriers that enable time- or stimulus-programmed drug release holds great promise to enhance the treatment efficacy in tumors. Here, hyaluronic acid (HA)-coated liposomes were designed to efficiently deliver multi-organelle-targeted and ALP/GSH dual-responsive prodrugs for combination therapy on colon tumors. In this system (designated CPTP/RA-HALipo), the unique natural cyclopeptide RA-V was linked covalently to a near-infrared (NIR) fluorophore through a disulfide linker, which was subsequently loaded in the cationic liposome core of CPTP/RA-HALipo, while the ALP-activatable phosphate CPT (CPTP) was encapsulated in the HA shell. In the tumor microenvironment, the HA shell of CPTP/RA-HALipo was partially degraded by HAase, thereby allowing the release of CPTP. The released phosphate prodrug CPTP was activated through hydrolysis of the phosphate esters by brush border-associated enzymes. The cationic liposome coated with the remaining HA could selectively enter CD44 overexpressed cells via receptor-mediated endocytosis into the lysosome, in which the acidic microenvironment degraded the liposomes to release the mitochondria-targeted theranostic agent RA-S-S-Cy. More significantly, the GSH-activatable NIR fluorescence of Cy5.5 made it possible to realize in vivo and in situ dynamic monitoring of drug release in a noninvasive manner. The organelle-specific and multi-stimuli responsive nanoparticles have shown precise control over drug delivery and release, leading to superior in vitro and/or in vivo anti-cancer efficacy. This approach represents a novel interactive drug delivery system that can synergistically differentiate the extracellular, cell membranal and intracellular targets to promote spatial and temporal control of drug release.

2.
Sci Total Environ ; 855: 158851, 2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36155047

RESUMEN

Accumulating evidences show that the hazardous substance atmospheric nanoplastics increase the respiratory risk of individuals, but the inside toxicity mechanisms to lung tissue remain unclear. This study aims at investigating the potential mechanisms of inhaled cationic polystyrene nanoplastics (amine-polystyrene nanoplastics, APS-NPs)-induced pulmonary toxicity on mice. In vivo, the mice intratracheal administrated with APS-NPs suspension (5 mg/kg) were found inflammatory infiltrates in lung tissues through histopathology analysis. Furthermore, transcriptome analysis demonstrated that 1821 differentially expressed mRNA between APS group and control group were dominantly associated with 288 known KEGG pathways, indicating that APS-NPs might cause early inflammatory responses in lung tissue by activating the NLRP3/capase-1/IL-1ß signaling pathway. Moreover, in vitro results also showed that NLRP3 inflammasome could be activated to induce pyroptosis in MLE-12 cells after exposure to APS-NPs. And, MH-S cells after exposure to APS-NPs exhibited increased Irg1 proteins, leading to the increasing generation of ROS and inflammatory factors (e.g., tnf-α, il-6, il-1ß). In conclusion, these results revealed that Irg1/NF-κB/NLRP3/Caspase-1 signaling pathway was activated significantly after exposing to APS-NPs, leading to pulmonary toxicity on mice. Intriguingly, prior administration of the clinical antioxidant N-acetylcysteine (NAC) could serve as a possible candidate for the prevention and treatment of pulmonary toxicity induced by APS-NPs. The study contributes to a better understanding of the potential risks of environmental nanoplastics to humans and its improvement measure.


Asunto(s)
Acetilcisteína , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Animales , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Acetilcisteína/farmacología , Antioxidantes , Poliestirenos/toxicidad , Microplásticos
3.
Acta Pharm Sin B ; 12(4): 2103-2119, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35847496

RESUMEN

Checkpoint blockade-based immunotherapy has shown unprecedented effect in cancer treatments, but its clinical implementation has been restricted by the low host antitumor response rate. Recently, chemotherapy is well recognized to activate the immune system during some chemotherapeutics-mediated tumor eradication. The enhancement of immune response during chemotherapy might further improve the therapeutic efficiency through the synergetic mechanism. Herein, a synergistic antitumor platform (designated as BMS/RA@CC-Liposome) was constructed by utilizing CT26 cancer-cell-biomimetic nanoparticles that combined chemotherapeutic drug (RA-V) and PD-1/PD-L1 blockade inhibitor (BMS-202) to remarkably enhance antitumor immunity. In this study, the cyclopeptide RA-V as chemotherapeutic drugs directly killing tumor cells and BMS-202 as anti-PD agents eliciting antitumor immune responses were co-encapsulated in a pH-sensitive nanosystem. To achieve the cell-specific targeting drug delivery, the combination therapy nanosystem was functionalized with cancer cell membrane camouflage. The biomimetic drug delivery system perfectly disguised as endogenous substances, and realized elongated blood circulation due to anti-phagocytosis capability. Moreover, the BMS/RA@CC-Liposome also achieved the selective targeting of CT26 cells by taking advantage of the inherent homologous adhesion property of tumor cells. The in vitro and in vivo experiments revealed that the BMS/RA@CC-Liposome realized PD-1/PD-L1 blockade-induced immune response, RA-V-induced PD-L1 down-regulation and apoptosis in cancer cells. Such a system combining the advantages of chemotherapy and checkpoint blockade-based immunotherapy to create an immunogenic tumor microenvironment systemically, demonstrated improved therapeutic efficacy against hypoxic tumor cells and offers an alternative strategy based on the immunology of the PD-1/PD-L1 pathway.

4.
Int J Nanomedicine ; 16: 4929-4942, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326635

RESUMEN

BACKGROUND: Natural cyclopeptide RA-XII, isolated from Rubia yunnanensis, is a promising chemotherapeutic agent for colon cancer. The photosensitizer protoporphyrin-IX attached with triphenylphosphonium (TPP) could possess mitochondria targeting capacity and exert photodynamic therapy (PDT) by inducing oxidizing damage to the mitochondria and cell apoptosis eventually. In this work, pH-sensitive liposomes were constructed to simultaneously deliver RA-XII as a chemotherapeutic drug and modified porphyrin as a mitochondria-targeting photosensitizer to treat colon cancer, and verified its mechanism of action and antitumor therapeutic efficacy. METHODS: The colon cancer targeting liposome nanoparticle RA/TPPP-Lip was synthesized using thin film hydration. The therapeutic effect and targeting ability of RA/TPPP-Lip was investigated in vitro. And use HCT116 cell allogeneic subcutaneous transplantation tumor model to investigate the anti-tumor and targeting effects of RA/TPPP-Lip in vivo. RESULTS: RA/TPPP-Lip gained the targeting ability through surface-modified HA to increase the accumulation of RA-XII and TPPP in colon cancer cells. A series of in vitro experimental results showed that TPPP produced cytotoxic ROS under laser irradiation to directly damage cell mitochondria and played a combined role with RA-XII, making RA/TPPP-Lip the best colon cancer cell growth inhibitory effect. Furthermore, in vivo antitumor experiments showed that the RA/TPPP-Lip substantially accumulated at the tumor site and efficiently repressed tumor growth in nude mice. CONCLUSION: We have successfully designed a new cancer-targeted nanomedicine platform (RA/TPPP-Lip) for the collaborative treatment of colon cancer, which can achieve the targeted continuous release of multiple therapeutic drugs. This work provides a new strategy for precise combination therapy, which may promote the further development of collaborative cancer treatment platforms.


Asunto(s)
Neoplasias del Colon , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Humanos , Ácido Hialurónico , Liposomas , Ratones , Ratones Desnudos , Mitocondrias , Péptidos Cíclicos , Fármacos Fotosensibilizantes/farmacología
5.
Int J Pharm ; 584: 119455, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32464235

RESUMEN

Chronic kidney disease (CKD) is one of the leading public health problems worldwide and finally progresses to end-stage renal disease. The therapeutic options of CKD are very limited. Thus, development of drug delivery systems specific-targeting to kidney may offer more options. Here we developed an efficient kidney-targeted drug delivery system using a FITC labeled renal tubular-targeting peptide modified PLGA-PEG nanoparticles and investigated the intrarenal distribution and cell-type binding. We found that the modified nanoparticles with an approximate diameter of 200 nm exhibited the highest binding capacity with HK-2 cells and fluorescence and immunohistochemical analysis showed they mainly localized in renal proximal tubules by passing through the basolateral side. Furthermore, these kidney-specific nanoparticles could significantly enhance the therapeutic effects of asiatic acid, an insoluble triterpenoid compound as drug delivery carriers. In conclusion, these results suggest the potential of the peptide modified PLGA-PEG nanoparticles as kidneytargeted drug delivery system to proximal tubular cells in treatment of CKD.


Asunto(s)
Sistemas de Liberación de Medicamentos , Enfermedades Renales/tratamiento farmacológico , Nanopartículas/administración & dosificación , Triterpenos Pentacíclicos/administración & dosificación , Péptidos/administración & dosificación , Poliésteres/administración & dosificación , Polietilenglicoles/administración & dosificación , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo III/metabolismo , Fluoresceína-5-Isotiocianato/administración & dosificación , Fluoresceína-5-Isotiocianato/farmacocinética , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/metabolismo , Masculino , Triterpenos Pentacíclicos/farmacocinética , Péptidos/farmacocinética , Poliésteres/farmacocinética , Polietilenglicoles/farmacocinética , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo
6.
Biomater Sci ; 8(1): 256-265, 2019 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-31687671

RESUMEN

Chemotherapy is a dominant treatment modality for different types and stages of cancer. However, hypoxia is one of the undesirable limitations of chemotherapy, which reduces the therapeutic efficiency in cancer treatment, ultimately leading to failure of the treatment. Herein, an ideal chemosensitization system capable of attenuating the tumor hypoxia microenvironment and enhancing chemotherapy effects in tumors was designed. This system (designated as the RA/RX Liposome) uses for the first time a pH-sensitive liposome to co-deliver cyclopeptide RA-V as chemotherapeutic drugs and antisense oligonucleotides as HIF-1α inhibitors (RX-0047) for attenuating tumor hypoxia, as well as a caspase-8 activation probe for therapeutic self-monitoring. After modification with death receptor 5-specific antibodies (anti-DR5) on the surface of the liposome, the RA/RX Liposome can successfully deliver components targeting colon tumors in vivo. This work should synergistically enhance the therapeutic effects of the treatment by successfully down-regulating HIF-1α expression against tumor hypoxia during the RA-V-induced apoptotic process. More importantly, the RA/RX Liposome can be precisely applied for therapeutic self-monitoring with the light-up fluorescence of the caspase-8 probe.


Asunto(s)
Anticuerpos/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Oligonucleótidos/administración & dosificación , Péptidos Cíclicos/administración & dosificación , Hipoxia Tumoral/efectos de los fármacos , Animales , Anticuerpos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Caspasa 8/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Sinergismo Farmacológico , Femenino , Células HCT116 , Células HT29 , Humanos , Liposomas , Ratones , Oligonucleótidos/farmacología , Péptidos Cíclicos/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Theranostics ; 9(1): 90-103, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662556

RESUMEN

Activatable theranostic agents, which combine fluorescent reporters with masked chemotherapeutic agents that are activated by tumor-associated stimuli, would be attractive candidates to improve the tumor selectivity of chemotherapy. This work reports a ROS/GSH dual-activatable and O2­evolving theranostic nanosystem (RA-S-S-Cy@PLGA NPs) for highly selective therapy against hypoxic tumors and in situ fluorescence-tracking of cancer chemotherapy. Methods: In this system, the newly designed theranostic agent (RA-S-S-Cy) is composed of a disulfide bond as a cleavable linker, a near infrared (NIR) active fluorophore as a fluorescent tracker, and a natural cyclopeptide RA-V as the active anti-cancer agent. Upon reaction with the high level of intracellular glutathione (GSH), disulfide cleavage occurs, resulting in concomitant active drug RA-V release and significant NIR fluorescence increase. To further improve the tumor targeting of RA-S-S-Cy and achieve redox dual-responsiveness, RA-S-S-Cy was incorporated into the c(RGDfK)-targeted PLGA nanoparticles together with an O2-generating agent (catalase) to produce RA-S-S-Cy@PLGA NPs. Results: The cell-specific and redox dual-activatable release of RA-V lead to enhanced therapeutic outcomes in vivo and in vitro. More significantly, the RA-S-S-Cy@PLGA NPs were successfully applied for monitoring of drug release and chemotherapeutic efficacy in situ by "turn-on" NIR fluorescence. Conclusions: RA-S-S-Cy@PLGA NPs would be efficient theranostic nanosystems for more precise therapy against hypoxic tumors and provides a potential tool for deeper understanding of drug release mechanisms.


Asunto(s)
Antineoplásicos/administración & dosificación , Quimioterapia/métodos , Hipoxia , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Imagen Óptica/métodos , Nanomedicina Teranóstica/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/administración & dosificación , Trasplante de Neoplasias , Neoplasias/patología , Oxidación-Reducción , Trasplante Heterólogo , Resultado del Tratamiento
8.
J Mater Chem B ; 6(26): 4351-4359, 2018 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32254510

RESUMEN

This work reports a dimeric BODIPY (BDP)-loaded liposome with conjugation of anti-HIF antibodies for dual hypoxia marker imaging and nitroreductase (NTR)-activatable photodynamic therapy (PDT) against hypoxic tumors. In this theranostic nanosystem (designated Ab-DiBDP NPs), the newly designed orthogonal BDP dimer has high 1O2 quantum yield, and the substitution of a nitro group at the meso-position leads to the NTR-controllable activation of phototoxicity and fluorescence. Both in vivo and in vitro experiments demonstrate that the NTR-activatable PDT liposome can efficiently destroy cancer cells and prevent damage to normal cells. More significantly, the fascinating advantage of the nanoprobe is the synergy between the Cy 7-marked anti-HIF-1α antibody and the NTR-activatable DiBDP, which significantly improves the accuracy of tumor hypoxia imaging by simultaneous detection of NTR and HIF-1α. Therefore, this work presents a new paradigm for NTR-triggered PDT against cancer cells and provides a new avenue for precise tumor hypoxia diagnosis.

9.
Theranostics ; 7(15): 3781-3793, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109776

RESUMEN

A programmed drug delivery system that can achieve sequential release of multiple therapeutics under different stimulus holds great promise to enhance the treatment efficacy and overcome multi-drug resistance (MDR) in tumor. Herein, multi-organelle-targeted and pH/ cytochrome c (Cyt c) dual-responsive nanoparticles were designed for combination therapy on resistant tumor. In this system (designated DGLipo NPs), doxorubicin (Dox) was intercalated into the DNA duplex containing a Cyt c aptamer, which subsequently loaded in the dendrigraftpoly-L-lysines (DGL) cores of DGLipo NPs, while cyclopeptide RA-V was doped into the pH-sensitive liposomal shells. After dual modification with c(RGDfK) and mitochondria-penetrating peptide (MPP), DGLipo NPs could successively deliver the two drugs into lysosome and mitochondria of cancer cells, and achieve sequential drug release in virtue of the unique characteristic of these two organelles. The organelle-specific and spatiotemporally controlled release of Dox and RA-V led to enhanced therapeutic outcomes in MDR tumor. More significantly, the DGLipo NPs were successfully applied to monitor Cyt c release during mitochondria-mediated apoptotic process. This work represents a versatile strategy for precise combination therapy against resistant tumor with spatiotemporal control, and provides a potential tool for Cyt c-related apoptotic studies.


Asunto(s)
Citocromos c/metabolismo , Doxorrubicina/uso terapéutico , Péptidos Cíclicos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Células HeLa , Humanos , Células MCF-7 , Ratones , Microscopía Electrónica de Transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA