Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Pharm Sin B ; 13(12): 4983-4998, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38045059

RESUMO

Lipid-coated microbubbles are widely used as an ultrasound contrast agent, as well as drug delivery carriers. However, the two main limitations in ultrasound diagnosis and drug delivery using microbubbles are the short half-life in the blood system, and the difficulty of surface modification of microbubbles for active targeting. The exosome, a type of extracellular vesicle, has a preferentially targeting ability for its original cell. In this study, exosome-fused microbubbles (Exo-MBs) were developed by embedding the exosome membrane proteins into microbubbles. As a result, the stability of Exo-MBs is improved over the conventional microbubbles. On the same principle that under the exposure of ultrasound, microbubbles are cavitated and self-assembled into nano-sized particles, and Exo-MBs are self-assembled into exosome membrane proteins-embedded nanoparticles (Exo-NPs). The Exo-NPs showed favorable targeting properties to their original cells. A photosensitizer, chlorin e6, was loaded into Exo-MBs to evaluate therapeutic efficacy as a drug carrier. Much higher therapeutic efficacy of photodynamic therapy was confirmed, followed by cancer immunotherapy from immunogenic cell death. We have therefore developed a novel ultrasound image-guided drug delivery platform that overcomes the shortcomings of the conventional ultrasound contrast agent and is capable of simultaneous photodynamic therapy and cancer immunotherapy.

2.
Small ; 19(47): e2301377, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37491793

RESUMO

Cancer immunotherapy is a promising therapy to treat cancer patients with minimal toxicity, but only a small fraction of patients responded to it as a monotherapy. In this study, a strategy to boost therapeutic efficacy by combining an immunotherapy based on ex vivo expanded tumor-reactive T cells is devised, or adoptive cell therapy (ACT), with photothermal therapy (PTT). Smart gold nanoparticles (sAuNPs), which aggregates to form gold nanoclusters in the cells, are loaded into T cells, and their photothermal effects within T cells are confirmed. When transferred into tumor-bearing mice, large number of sAuNP-carrying T cells successfully infiltrate into tumor tissues and exert anti-tumor activity to suspend tumor growth, but over time tumor cells evade and regrow. Of note, ≈20% of injected doses of sAuNPs are deposited in tumor tissues, suggesting T cells are an efficient nanoparticle tumor delivery vehicle. When T cells no longer control tumor growth, PTT is performed to further eliminate tumors. In this manner, ACT and PTT are temporally coupled, and the combined immuno-photothermal treatment demonstrated significantly greater therapeutic efficacy than the monotherapy.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias , Humanos , Animais , Camundongos , Ouro/uso terapêutico , Linfócitos T , Nanopartículas Metálicas/uso terapêutico , Neoplasias/tratamento farmacológico , Terapia Combinada , Fototerapia , Linhagem Celular Tumoral
3.
Biomater Sci ; 11(13): 4537-4548, 2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-37158297

RESUMO

For the successful development of various cellular products in cancer immunotherapy, an effective ex vivo priming technique for immune cells is often required. Among a variety of immunomodulatory substances, tumor cell lysates (TCLs) have been considered a robust immune activator with high adjuvanticity and tumor antigen population. Therefore, the present study suggests a novel ex vivo dendritic cell (DC) priming technique that utilizes (1) squaric acid (SqA)-mediated oxidation of source tumor cells to obtain antigenic TCLs with an increased immunogenic potential and (2) a coacervate (Coa) colloidal complex as an exogenous TCL carrier. Elevated oxidation by SqA-treated source tumor cells resulted in an increased immunogenic potential, indicated by a high level of damage-associated molecular pattern molecules in TCLs that could sufficiently stimulate DCs. Moreover, to effectively deliver these exogenous immunomodulating TCL DCs, Coa (i.e., a colloidal micro-carrier using cationic mPEGylated poly(ethylene arginyl aspartate diglyceride) and anionic heparin) was utilized for the sustained release of cargo TCLs and for preserving their bioactivity. Coa-mediated ex vivo delivery of SqA-treated TCLs (SqA-TCL-Coa) effectively promoted DC maturation through the enhanced uptake of antigens into target DCs, increased expression of DC activation markers, facilitated secretion of pro-inflammatory cytokines from activated DCs, and improved major histocompatibility complex-I dependent cross-presentation of a colorectal cancer specific antigen. Therefore, based on antigenic and adjuvant behaviors, our Coa-mediated exogenous delivery of SqA-TCL could be a promising application as a facile ex vivo DC priming strategy for further cell-based cancer immunotherapies.


Assuntos
Neoplasias , Humanos , Neoplasias/metabolismo , Antígenos de Neoplasias , Apresentação Cruzada , Citocinas/metabolismo , Células Dendríticas
4.
Mater Horiz ; 10(6): 2215-2225, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37000519

RESUMO

In this work, we describe the development of an implantable ionic device that can deliver a spatially targeted light source to tumor tissues in a controllable manner. The motivation behind our approach is to overcome certain limitations of conventional approaches where light is delivered from the outside of the body and only achieves low penetration depths. Also, to avoid the issues that come from the periodic need to replace the device's battery, we utilize a wireless power transfer system synchronized with light operation in an implantable structure. In our testing of this implanted, soft ionic, gel-based device that receives power wirelessly, we were able to clearly observe its capability to effectively deliver light in a harmonious and stable configuration to adjacent tissues. This approach reduces the mechanical inconsistencies seen in conventional systems that are induced by mismatches between the mechanical strength of conventional metallic components and that of biological tissues. The light delivering performance of our device was studied in depth under the various conditions set by adjusting the area of the gel receivers, the ion concentration and the ion types used in the gel components. The enhanced antitumor effects of our device were observed through in vitro cell tests, in comparison with treatments using the conventional approach of using direct light from outside the body. Full encapsulation using biocompatible elastomers enables our device to provide good functional stability, while implantation for about 3 weeks in the in vivo model showed the effective targeted photodynamic treatments made possible by our approach. Our advanced approach of designing the implantable platform based on ionic gel components allows us to iteratively irradiate a target with light whenever required, making the technology particularly suited to long-term treatment of residual tumors while facilitating further practical and clinical development.


Assuntos
Fotoquimioterapia , Tecnologia sem Fio , Próteses e Implantes , Fontes de Energia Elétrica , Tecnologia
5.
ACS Sens ; 6(3): 693-697, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33606518

RESUMO

Natural killer (NK) cells are a subset of innate lymphoid cells playing an important role in immune surveillance and early defense against infection and cancer. They recognize and directly kill infected or transformed cells. At the same time, they produce various cytokines and chemokines to regulate other immune cells. NK cell activity can be a useful marker for health screenings because impaired NK cell functions may indicate a more susceptible environment for infection or tumor development. Currently, most NK cell activity assays are focused on measuring either cytokine secretion, in particular, interferon γ (IFN-γ), or cytotoxicity against target cells such as K562, thus only providing partial information on NK cell activity. In order to develop a comprehensive test for measuring NK cell function, cytotoxicity and cytokine secretion ability should be measured simultaneously. In addition, current NK cell assays are performed by stimulating NK cells with cocktails of cytokines, antibody-coated beads, or live target cells. In this study, we developed multifunctional microparticles for NK cell activity assay (MNAs) that allow simultaneous stimulation and sensing various NK cell activities, including cytokine secretion and cytotoxicity. The surfaces of MNAs are decorated with multiple functional biomolecules, including antibodies that stimulate NK cells by engaging NK cell activating receptors, antibodies that can capture cytokines secreted by NK cells, and a peptide sensor that reacts with granzyme B, a key molecule released by NK cells for cytotoxicity. The performances of MNAs are assessed using flow cytometry and live cell imaging. NK cell activity is measured by simply mixing MNAs with NK cells and performing flow cytometry, and the results are comparable to those measured by standard NK cell activity assays.


Assuntos
Imunidade Inata , Células Matadoras Naturais , Citocinas , Citometria de Fluxo , Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...