Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biomacromolecules ; 25(2): 964-974, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38232296

RESUMEN

Thermosensitive nanoparticles can be activated by externally applying heat, either through laser irradiation or magnetic fields, to trigger the release of drug payloads. This controlled release mechanism ensures that drugs are specifically released at the tumor site, maximizing their effectiveness while minimizing systemic toxicity and adverse effects. However, its efficacy is limited by the low concentration of drugs at action sites, which is caused by no specific target to tumor sties. Herein, hyaluronic acid (HA), a gooey, slippery substance with CD44-targeting ability, was conjugated with a thermosensitive polymer poly(acrylamide-co-acrylonitrile) to produce tumor-targeting and thermosensitive polymeric nanocarrier (HA-P) with an upper critical solution temperature (UCST) at 45 °C, which further coloaded chemo-drug doxorubicin (DOX) and photosensitizer Indocyanine green (ICG) to prepare thermosensitive nanoreactors HA-P/DOX&ICG. With photosensitizer ICG acting as the "temperature control element", HA-P/DOX&ICG nanoparticles can respond to temperature changes when receiving near-infrared irradiation and realize subsequent structure depolymerization for burst drug release when the ambient temperature was above 45 °C, achieving programmable and on-demand drug release for effective antitumor therapy. Tumor inhibition rate increased from 61.8 to 95.9% after laser irradiation. Furthermore, the prepared HA-P/DOX&ICG nanoparticles possess imaging properties, with ICG acting as a probe, enabling real-time monitoring of drug distribution and therapeutic response, facilitating precise treatment evaluation. These results provide enlightenment for the design of active tumor targeting and NIR-triggered programmable and on-demand drug release of thermosensitive nanoreactors for tumor therapy.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Humanos , Fármacos Fotosensibilizantes/uso terapéutico , Hipertermia Inducida/métodos , Fototerapia/métodos , Doxorrubicina/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Verde de Indocianina/farmacología , Verde de Indocianina/química , Nanotecnología , Liberación de Fármacos , Línea Celular Tumoral
2.
Mol Pharm ; 18(11): 3966-3978, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34579532

RESUMEN

Stemness and metastasis are the two main challenges in cancer therapy and are related to disease relapse post-treatment. They both have a strong correlation with chemoresistance and poor prognosis, ultimately leading to treatment failure. It has been reported that chemotherapy can induce stemness and metastasis in many cancer types, especially treatment with the chemotherapeutic agent doxorubicin (DOX) in breast cancer. A combination treatment is an efficient and elegant approach in cancer therapy through simultaneous delivery of two or more drugs with a delivery system for its synergistic effect, which is not an additive of two individual drugs. Herein, we report a combinatorial system with DOX and all-trans retinoic acid (ATRA) to address both of the above issues. As a common critical regulatory factor for oncogenic signal transduction pathways, Pin1 is a specific isomerase highly expressed within various tumor cells. ATRA, a newly identified Pin1 inhibitor, can abolish several oncogenic pathways by effectively inhibiting and degrading overexpressed Pin1. We successfully developed a folic acid (FA)-modified chitosan (CSO)-derived polymer (FA-CSOSA) and obtained FA-CSOSA/DOX and FA-CSOSA/ATRA drug-loaded micelles. FA modification can improve the uptake of the nanoparticles in tumor cells and tumor sites via folate receptor-mediated cell internalization. Compared to treatment with DOX alone, the combined treatment induced 4T1 cell apoptosis in a synergistic manner. Reduced stemness-related protein expression and inhibited metastasis were observed during treatment with FA-CSOSA/DOX and FA-CSOSA/ATRA and were found to be associated with Pin1. Further in vivo experiments showed that treatment with FA-CSOSA/DOX and FA-CSOSA/ATRA resulted in 85.5% tumor inhibition, which was 2.5-fold greater than that of cells treated with DOX·HCl alone. This work presents a new paradigm for addressing chemotherapy-induced side effects via degradation of Pin1 induced by tumor-targeted delivery of DOX and ATRA.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Sistema de Administración de Fármacos con Nanopartículas/química , Tretinoina/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quitosano/química , Modelos Animales de Enfermedad , Doxorrubicina/farmacocinética , Liberación de Fármacos , Sinergismo Farmacológico , Femenino , Ácido Fólico/química , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Micelas , Peptidilprolil Isomerasa de Interacción con NIMA/antagonistas & inhibidores , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Distribución Tisular , Tretinoina/farmacocinética
3.
Biomaterials ; 275: 120958, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34130142

RESUMEN

Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) play an important role in the development of tumors by secreting a variety of cytokines or directly communicating with tumor cells, making TAMs-targeted therapeutic strategies very attractive. It has been reported that oncogene c-Myc is related to every aspect of the oncogenic process of tumor cells and the alternative activation of macrophages. Hence, we constructed a glycolipid nanocarrier containing ROS-responsive peroxalate linkages (CSOPOSA) for ROS-triggered release of drugs and further modified it with Ex 26 (Ex 26-CSOPOSA), a selective sphingosine 1-phosphate receptor 1 (S1PR1) antagonist, to achieve the dual-targeted delivery of the c-Myc inhibitor JQ1 via S1PR1, which is overexpressed on both tumor cells and TAMs, thereby inducing apoptosis of tumor cells, and blocking M2 polarization of macrophages. More strikingly, our studies found that JQ1 could effectively inhibit the migration of tumor cells induced by M2 macrophages-derived exosomes via blocking Caveolin-1 related intercellular exosome exchange through lncRNA H19 and miR-107. The in vivo results revealed that this dual-targeted delivery strategy effectively inhibited tumor growth and metastasis with less systemic toxicity, providing a potential method for effective tumor treatment.


Asunto(s)
Caveolina 1 , Exosomas , Neoplasias Experimentales/tratamiento farmacológico , Especies Reactivas de Oxígeno , Animales , Línea Celular Tumoral , Portadores de Fármacos , Liberación de Fármacos , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-myc , Células RAW 264.7 , Microambiente Tumoral
4.
Biomater Sci ; 8(21): 5955-5968, 2020 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-32966382

RESUMEN

The active targeting strategy has achieved inspiring progress for drug accumulation in tumor therapy; however, the insufficient expression level of many potential receptors poses challenges for drug delivery. Poly-γ-glutamic acid (γ-pGluA), a naturally occurring anionic biopolymer, showed high affinity with tumor-associated gamma-glutamyl transpeptidase (GGT), which localized on the cell surface and exhibited intracellular redox homeostasis-dependent expression pattern; thus, GGT was utilized for mediating endocytosis of nanoparticles. Herein, GGT-targeting nanopolyplexes (γ-pGluA-CSO@Fe3+, PCFN) consisting of cationic chitosan and GGT-targeting γ-pGluA blended with iron ion were constructed to load reactive oxygen species-induced menadione (MA) and doxorubicin, which were utilized to investigate the mechanism of GGT up-regulation. Briefly, the pretreated PCFN/MA induced an intracellular oxidative stress environment, which facilitated adjusted up-regulated GGT expression and boosted tumor targeting. Subsequently, the destroyed redox homeostasis sensitized tumors for synergistic therapy. The innovative strategy of augmenting active targeting by disturbing intracellular redox homeostasis offers insight for the application of γ-pGluA-derived nanopolyplexes.


Asunto(s)
Ácido Glutámico , gamma-Glutamiltransferasa , Homeostasis , Oxidación-Reducción , Regulación hacia Arriba , gamma-Glutamiltransferasa/genética , gamma-Glutamiltransferasa/metabolismo
5.
Int J Nanomedicine ; 15: 2717-2732, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32368051

RESUMEN

BACKGROUND: Phototherapy is a potential new candidate for glioblastoma (GBM) treatment. However inadequate phototherapy due to stability of the photosensitizer and low target specificity induces the proliferation of neovascular endothelial cells for angiogenesis and causes poor prognosis. METHODS: In this study, we constructed c(RGDfk)-modified glycolipid-like micelles (cRGD-CSOSA) encapsulating indocyanine green (ICG) for dual-targeting neovascular endothelial cells and tumor cells, and cRGD-CSOSA/ICG mediated dual effect of PDT/PTT with NIR irradiation. RESULTS: In vitro, cRGD-CSOSA/ICG inhibited cell proliferation and blocked angiogenesis with NIR irradiation. In vivo, cRGD-CSOSA/ICG exhibited increased accumulation in neovascular endothelial cells and tumor cells. Compared with that of CSOSA, the accumulation of cRGD-CSOSA in tumor tissue was further improved after dual-targeted phototherapy pretreatment. With NIR irradiation, the tumor-inhibition rate of cRGD-CSOSA/ICG was 80.00%, significantly higher than that of ICG (9.08%) and CSOSA/ICG (42.42%). Histological evaluation showed that the tumor vessels were reduced and that the apoptosis of tumor cells increased in the cRGD-CSOSA/ICG group with NIR irradiation. CONCLUSION: The cRGD-CSOSA/ICG nanoparticle-mediated dual-targeting phototherapy could enhance drug delivery to neovascular endothelial cells and tumor cells for anti-angiogenesis and improve the phototherapy effect of glioblastoma, providing a new strategy for glioblastoma treatment.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/terapia , Verde de Indocianina/administración & dosificación , Nanopartículas/administración & dosificación , Neovascularización Patológica/tratamiento farmacológico , Fototerapia/métodos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glioblastoma/patología , Glucolípidos/química , Humanos , Verde de Indocianina/química , Ratones Desnudos , Micelas , Nanopartículas/química , Oligopéptidos/química , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/farmacología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Carbohydr Polym ; 240: 116270, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32475559

RESUMEN

Tumor-draining lymph node (TDLN), already bathed in tumor antigens, has been proposed as an intriguing site for cancer immunotherapy. Targeted delivery of adjuvants to TDLN, presumably could induce antitumor immunity for personalized immunotherapy. Although molecular adjuvants can be used for personalized immunotherapy, their efficacy is limited by insufficient antigen uptake by dendritic cells (DCs). In contrast, nanomaterial-based adjuvants can enhance antigen uptake by DCs by capturing antigens. Herein, mannose modified stearic acid-grafted chitosan micelles (MChSA), which presumably could target TDLN, were engineered to capture endogenous antigens and enhance antigen uptake by DCs for personalized immunotherapy. MChSA micelles showed strong antigen-capturing and TDLN targeting ability. Importantly, MChSA micelles induced robust CD4+ and CD8+ T cell responses, stimulated antitumor related cytokine secretion and notably inhibited tumor growth. MChSA micelles, which can target TDLN to induce potent antitumor immune responses as antigen-capturing adjuvants, exhibit great potential in personalized cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Quitosano/química , Células Dendríticas/efectos de los fármacos , Ganglios Linfáticos , Neoplasias/terapia , Animales , Línea Celular Tumoral , Células Dendríticas/citología , Inmunoterapia , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Micelas
7.
RSC Adv ; 10(35): 20445-20459, 2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-35517758

RESUMEN

An insufficient drug concentration at the target site and drug efflux resulting in poor efficacy are recognized as important obstacles in osteoporosis treatment. Simvastatin (SIM), which can treat osteoporosis by promoting osteoblast differentiation and mineralization through the bone morphogenetic proteins (BMP)-Smad signaling pathway, has lower bioavailability, and less bone tissue distribution. Herein, novel lipid nanoparticles (LNPs) delivering SIM (SIM/LNPs) for osteoporosis therapy were developed with aspartic oligopeptide (ASP n , here ASP6)-based bone-targeting moieties grafted to the nanoparticles (SIM/ASP6-LNPs) in an attempt to increase the concentration of SIM in bones with a relatively low dose to minimize adverse effects. In vivo experiments indicated that the ASP6-LNPs exhibited ideal bone-targeting characteristics, and in vitro cell evaluation experiments showed LNPs have good biocompatibility with MC3T3-E1 cells. The cell mineralization experiment revealed that the SIM-loaded LNPs induced osteoblast differentiation and the formation of mineralized nodules in MC3T3-E1 cells, achieving the same efficacy as that of SIM. Pharmacodynamic experiments revealed that SIM/ASP6-LNPs improved the efficacy of SIM on the recovery of bone mineral density when compared to SIM/LNPs or to SIM alone. Therefore, SIM/ASP6-LNPs may represent a potential bone-targeting drug delivery system (DDS) that contributes to the development of a novel osteoporosis treatment.

9.
Theranostics ; 9(23): 6764-6779, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31660067

RESUMEN

Rationale: Nano-carrier based combinational therapies for tumor cells hold great potential to improve the outcomes of patients. However, cancer associated fibroblasts (CAFs) in desmoplastic tumors and the derived pathological tumor stroma severely impede the access and sensitibity of tumor cells to antitumor therapies. Methods: Glycolipid-based polymeric micelles (GLPM) were developed to encapsulate an angiotensin II receptor I inhibitor (telmisartan, Tel) and a cytotoxic drug (doxorubicin, DOX) respectively, which could exert combinational antitumor efficacy by reprogramming tumor microenvironment to expose the vulnerability of internal tumor cells. Results: As demonstrated, α-SMA positive CAFs significantly decreased after the pre-administration of GLPM/Tel in vitro, which accordingly inhibited the secretion of the CAFs derived stroma. The tumor vessels were further decompressed as a result of the alleviated solid stress inside the tumor masses, which promoted more intratumoral drug delivery and penetration. Ultimately, staged administration of the combined GLPM/Tel and GLPM/DOX at the screened molar ratio not only inhibited the stroma continuously, but also achieved a synergistic antitumor effect through the apoptosis-related peroxisome proliferator-activated receptor-gamma (PPAR-γ) pathway. Conclusion: In summary, the strategy of suppressing tumor stroma for subsequent combinational therapies against internal breast tumor cells could provide avenues for management of intractable desmoplastic tumors.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de los fármacos , Doxorrubicina/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Micelas , Telmisartán/farmacología , Microambiente Tumoral/efectos de los fármacos , Células 3T3 , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Fibroblastos Asociados al Cáncer/metabolismo , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Femenino , Glucolípidos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , PPAR gamma/metabolismo , Telmisartán/administración & dosificación , Telmisartán/uso terapéutico
10.
Int J Pharm ; 555: 303-313, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30471372

RESUMEN

Multi-cycle treatment strategies were frequently applied in anti-tumor therapy in clinic. However, numerous tumors developed drug resistance during this process, and few researches paid attention to the multi-cycle treatment process when a nano carrier was adopted. In this research, a glycolipid-like nanocarrier encapsulating anti-tumor drug doxorubicin (DOX) was adopted to perform a long term drug stimulation in vitro cell line and a tri-cycle treatment on xenograft tumors to explore its effect in process of developing drug resistance. As expected, tumors treated by free doxorubicin hydrochloride (DOX·HCl) showed obvious increase of P-glycoprotein, while for tumors treated by nanocarrier encapsulated doxorubicin, the P-glycoprotein level stayed low as untreated group. Further exploration found that MDR1 gene transcription got involved in the resistance induction mechanism as its mRNA levels in DOX·HCl stimulated cells were thousands of times of those in parent cell. It concluded that tri-cycle therapy with the glycolipid-like nanocarrier would not result in acquired drug resistance. These results also implied that nano-drug delivery possessed ability in avoiding acquiring drug resistance ability during long-term treatment which may have potential use in clinic.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Nanopartículas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quitosano/química , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Femenino , Glucolípidos/química , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA