Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nanoscale Horiz ; 8(8): 1062-1072, 2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37306000

RESUMO

One main obstacle to targeted cancer therapies is the immunosuppressive tumor microenvironment, which can facilitate tumor growth and induce resistance to antitumor treatments. Recent studies have indicated that treatment combined with immunotherapy often yields a better prognosis than monotherapy. Bacterial membrane vesicles (MVs), nanostructures released from the membrane of bacteria, can be used as natural nanocarriers for drug delivery and stimulate an immune response because of their immunogenicity. Inspired by the development of synergistic therapeutic strategies, we herein propose a novel nanovaccine-based platform to achieve chemotherapy, ferroptosis therapy, and immunotherapy simultaneously. By simply culturing magnetotactic bacteria in the medium with doxorubicin (DOX) and then extracting specialized MVs (BMVs), BMV@DOX, which are membrane vesicles containing iron ions and DOX, were obtained. We confirmed that in BMV@DOX, the BMV component can stimulate the innate immune system, DOX acts as the chemotherapeutic agent and iron ions will induce ferroptosis. Furthermore, BMV@DOX vesicles modified with DSPE-PEG-cRGD peptides (T-BMV@DOX) have minimized systemic toxicity and increased tumor-specificity. We demonstrated that the smart MVs-based nanovaccine system not only showed superior performance in the treatment of 4T1 breast cancer but also effectively restrained the growth of drug-resistant MCF-7/ADR tumors in mice. Moreover, the nanovaccine could abrogate in vivo lung metastasis of tumor cells in a 4T1-Luc cell induced-lung breast cancer metastasis model. Collectively, the MVs-based nanoplatform offers an alternative promise for surmounting the limitations of monotherapy and may deserve further study for application in synergistic cancer therapy.


Assuntos
Ferroptose , Neoplasias , Animais , Camundongos , Doxorrubicina/uso terapêutico , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Imunoterapia , Neoplasias/tratamento farmacológico , Microambiente Tumoral
2.
Adv Healthc Mater ; 11(14): e2200841, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35579102

RESUMO

Nanomaterials as T1 /T2 dual-mode magnetic resonance imaging (MRI) contrast agents have great potential in improving the accuracy of tumor diagnosis. Applications of such materials, however, are limited by the complicated chemical synthesis process and potential biosafety issues. In this study, the biosynthesis of manganese (Mn)-doped magnetosomes (MagMn) that not only can be used in T1 /T2 dual-mode MR imaging with self-confirmation for tumor detection, but also improve the photothermal conversion efficiency for MRI-guided photothermal therapy (PTT) is reported. The MagMn nanoparticles (NPs) are naturally produced through the biomineralization of magnetotactic bacteria by doping Mn into the ferromagnetic iron oxide crystals. In vitro and in vivo studies demonstrated that targeting peptides functionalized MagMn enhanced both T1 and T2 MRI signals in tumor tissue and significantly inhibited tumor growth by the further MRI-guided PTT. It is envisioned that the biosynthesized multifunctional MagMn nanoplatform may serve as a potential theranostic agent for cancer diagnosis and treatment.


Assuntos
Magnetossomos , Nanopartículas , Neoplasias , Meios de Contraste/química , Humanos , Imageamento por Ressonância Magnética/métodos , Manganês , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Terapia Fototérmica , Nanomedicina Teranóstica/métodos
3.
Nanoscale ; 12(25): 13513-13522, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32555818

RESUMO

Tumor-targeted drug carriers are becoming attractive for precise drug delivery in anti-tumor therapy. However, a lot of the reported drug delivery systems are complicatedly designed and their destiny in vivo is beyond our control, which limited their clinical applications. Hence, it is urgently needed to develop spatio-manipulable self-propelled nanosystems for drug delivery in a facile way. Here, we have successfully constructed drug-internalized bacterial swimmers, whose movement can be manually controlled by an external magnetic field (MF). We demonstrate that the swimmers maintain the mobility to align and swim along MF lines. Further studies reveal that the doxorubicin (DOX-) internalized bacterial swimmers are able to navigate toward tumor sites under the guidance of MF, rendering enhanced anti-tumor efficacy compared with that of dead ones and free DOX. Therefore, the MF-guided bacterial swimmers hold great promise for spatio-manipulable drug delivery in precision medicine.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Portadores de Fármacos , Humanos , Campos Magnéticos , Neoplasias/tratamento farmacológico
4.
Biomolecules ; 9(5)2019 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-31060320

RESUMO

Improving the efficacy of nanoparticles (NPs) delivery to tumors is critical for cancer diagnosis and therapy. In our previous work, amphiphilic peptide APPA self-assembled nanocarriers were designed and constructed for cargo delivery to tumors with high efficiency. In this study, we explore the use of APPA self-assembled peptosomes as a nanoparticle adjuvant to enhance the delivery of nanoparticles and antibodies to integrin αvß3 and neuropilin-1 (NRP1) positive tumors. The enhanced tumor delivery of coadministered NPs was confirmed by better magnetosome (Mag)-based T2-weighted magnetic resonance imaging (MRI), liposome-based fluorescence imaging, as well as the improved anti-tumor efficacy of monoclonal antibodies (trastuzumab in this case) and doxorubicin (DOX)-containing liposomes. Interestingly, the improvement is most significant for the delivering of compounds that have active or passive tumor targeting ability, such as antibodies or NPs that have enhanced permeability and retention (EPR) effect. However, for non-targeting small molecules, the effect is not significant. In vitro and in vivo studies suggest that both peptosomes and the coadministered compounds might be internalized into cells through a NRP1 mediated co-endocytosis (CoE) pathway. The improved delivery of coadministered NPs and antibodies to tumors suggests that the coadministration with APPA self-assembled peptosomes could be a valuable approach for advancing αvß3 and NRP1 positive tumors diagnosis and therapy.


Assuntos
Sistemas de Liberação de Medicamentos , Endocitose , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Neuropilina-1/metabolismo , Peptídeos/administração & dosagem , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Endocitose/efeitos dos fármacos , Feminino , Humanos , Imageamento por Ressonância Magnética , Magnetossomos , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...