Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Clin Ultrasound ; 52(1): 68-77, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37907965

RESUMEN

Due to lymphocytic infiltration of the salivary and lacrimal glands, Sjogren's syndrome (SS), a systemic autoimmune illness that mostly affects the exocrine glands, causes dry mouth (xerostomia) and dry eyes (xerophthalmia). Additionally, SS is associated with various comorbidities such as cardiovascular diseases, infections, musculoskeletal diseases, and cancers. Among patients with SS, xerophthalmia frequently arises as a complication, leading to insufficient tear production or rapid tear evaporation, thereby causing discomfort, irritation, and a gritty sensation in the eyes. This article aims to examine recent advancements in the imaging of the lacrimal gland in Sjögren's syndrome and briefly discusses the utilization of various imaging examinations for the lacrimal gland in this particular disease.


Asunto(s)
Aparato Lagrimal , Síndrome de Sjögren , Xeroftalmia , Xerostomía , Humanos , Síndrome de Sjögren/complicaciones , Síndrome de Sjögren/diagnóstico por imagen , Aparato Lagrimal/diagnóstico por imagen , Diagnóstico por Imagen
2.
J Nanobiotechnology ; 20(1): 213, 2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35524280

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with pathophysiological characteristics of transforming growth factor-ß (TGF-ß), and reactive oxygen species (ROS)-induced excessive fibroblast-to-myofibroblast transition and extracellular matrix deposition. Macrophages are closely involved in the development of fibrosis. Nuclear factor erythroid 2 related factor 2 (Nrf2) is a key molecule regulating ROS and TGF-ß expression. Therefore, Nrf2 signaling modulation might be a promising therapy for fibrosis. The inhalation-based drug delivery can reduce systemic side effects and improve therapeutic effects, and is currently receiving increasing attention, but direct inhaled drugs are easily cleared and difficult to exert their efficacy. Therefore, we aimed to design a ROS-responsive liposome for the Nrf2 agonist dimethyl fumarate (DMF) delivery in the fibrotic lung. Moreover, we explored its therapeutic effect on pulmonary fibrosis and macrophage activation. RESULTS: We synthesized DMF-loaded ROS-responsive DSPE-TK-PEG@DMF liposomes (DTP@DMF NPs). DTP@DMF NPs had suitable size and negative zeta potential and excellent capability to rapidly release DMF in a high-ROS environment. We found that macrophage accumulation and polarization were closely related to fibrosis development, while DTP@DMF NPs could attenuate macrophage activity and fibrosis in mice. RAW264.7 and NIH-3T3 cells coculture revealed that DTP@DMF NPs could promote Nrf2 and downstream heme oxygenase-1 (HO-1) expression and suppress TGF-ß and ROS production in macrophages, thereby reducing fibroblast-to-myofibroblast transition and collagen production by NIH-3T3 cells. In vivo experiments confirmed the above findings. Compared with direct DMF instillation, DTP@DMF NPs treatment presented enhanced antifibrotic effect. DTP@DMF NPs also had a prolonged residence time in the lung as well as excellent biocompatibility. CONCLUSIONS: DTP@DMF NPs can reduce macrophage-mediated fibroblast-to-myofibroblast transition and extracellular matrix deposition to attenuate lung fibrosis by upregulating Nrf2 signaling. This ROS-responsive liposome is clinically promising as an ideal delivery system for inhaled drug delivery.


Asunto(s)
Fibrosis Pulmonar Idiopática , Factor 2 Relacionado con NF-E2 , Animales , Fibrosis , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Liposomas , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
3.
J Nanobiotechnology ; 19(1): 432, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930301

RESUMEN

BACKGROUND: Distant metastasis to vital organs is the major contributor to breast cancer mortality, and regional lymph node metastasis is an important facilitator of distant metastasis and recurrence in this cancer. The early diagnosis and precise treatment of lymph node metastasis are crucial for staging and prognosis in breast cancer. Herein, we report a visualized precision medicine nanoplatform of metastatic lymph nodes for ultrasonic/photoacoustic (US/PA) dual modal imaging-guided in situ targeted hyperthermia-combined chemotherapy. RESULTS: Carbon nanoparticles (CNs), approved by the China Food and Drug Administration, were loaded with docetaxel and rationally combined with anti-hypoxia-inducible factor 1α antibody-modified poly (lactic-co-glycolic acid) (PLGA) nanoparticles to achieve the combination of passive targeting at the lymph nodes and intracellular targeting at HIF 1α factor. The accumulation and retention of nanoparticles in metastatic lymph nodes via lymphatic delivery were enhanced. Docetaxel could be effectively offloaded by CNs that have active carbon nanoparticles, and the PLGA membrane prevented drug leakage. The nanoparticles exhibited excellent photothermal performance with a photothermal conversion efficiency of 28.9%, killing tumor cells in metastatic lymph nodes through hyperthermia. In vitro and in vivo systematic evaluations revealed that hyperpyrexia triggered the rupture of nanoparticles caused by the phase transition of perfluorohexane, resulting in docetaxel release for achieving in situ hyperthermia-combined chemotherapy. CONCLUSIONS: The laser-triggered highly efficient in situ chemotherapy nanosystem achieves targeted synergistic chemo-hyperthermia treatment of metastatic lymph nodes, and lymphatic delivery represents a strategy to avoid additional injury caused by drugs entering the blood circulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Hipertermia Inducida/métodos , Ganglios Linfáticos/metabolismo , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos/química , Anticuerpos/inmunología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carbono/química , Línea Celular Tumoral , Docetaxel/química , Docetaxel/metabolismo , Docetaxel/farmacología , Docetaxel/uso terapéutico , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Rayos Infrarrojos , Metástasis Linfática , Nanomedicina , Nanopartículas/metabolismo , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratas , Trasplante Heterólogo
4.
J Nanobiotechnology ; 19(1): 449, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952587

RESUMEN

BACKGROUND: Mono-therapeutic modality has limitations in combating metastatic lesions with complications. Although emerging immunotherapy exhibits preliminary success, solid tumors are usually immunosuppressive, leading to ineffective antitumor immune responses and immunotherapeutic resistance. The rational combination of several therapeutic modalities may potentially become a new therapeutic strategy to effectively combat cancer. RESULTS: Poly lactic-co-glycolic acid (PLGA, 50 mg) nanospheres were constructed with photothermal transduction agents (PTAs)-Prussian blue (PB, 2.98 mg) encapsulated in the core and chemotherapeutic docetaxel (DTX, 4.18 mg)/ immune adjuvant-imiquimod (R837, 1.57 mg) loaded in the shell. Tumor cell membranes were further coated outside PLGA nanospheres (designated "M@P-PDR"), which acted as "Nano-targeted cells" to actively accumulate in tumor sites, and were guided/monitored by photoacoustic (PA)/ magnetic resonance (MR) imaging. Upon laser irradiation, photothermal effects were triggered. Combined with DTX, PTT induced in situ tumor eradication. Assisted by the immune adjuvant R837, the maturation rate of DCs increased by 4.34-fold compared with that of the control. In addition, DTX polarized M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, relieving the immunosuppressive TME. The proportion of M2-TAMs decreased from 68.57% to 32.80%, and the proportion of M1-TAMs increased from 37.02% to 70.81%. Integrating the above processes, the infiltration of cytotoxic T lymphocytes (CTLs) increased from 17.33% (control) to 35.5%. Primary tumors and metastasis were significantly inhibited when treated with "Nano-targeted cells"-based cocktail therapy. CONCLUSION: "Nano-targeted cells"-based therapeutic cocktail therapy is a promising approach to promote tumor regression and counter metastasis/recurrence.


Asunto(s)
Antineoplásicos/uso terapéutico , Membrana Celular/química , Docetaxel/química , Nanopartículas/química , Neoplasias/terapia , Adyuvantes Inmunológicos/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Docetaxel/farmacología , Docetaxel/uso terapéutico , Ferrocianuros/química , Ferrocianuros/farmacología , Ferrocianuros/uso terapéutico , Humanos , Imiquimod/química , Imiquimod/inmunología , Inmunoterapia/métodos , Rayos Infrarrojos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Desnudos , Neoplasias/diagnóstico por imagen , Imagen Óptica , Terapia Fototérmica/métodos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
5.
Pharm Dev Technol ; 25(4): 454-463, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31873051

RESUMEN

Ultrasound can promote the drug release from drug-loaded substances and alter the tumor local microenvironment to facilitate the transport of drug carriers into the tumor tissues. Based on the altered tumor microenvironment, nanobubbles (NBs) as drug carriers with surfaces functionalized with targeting ligands can reach the tumor sites, thereby increasing the efficacy of chemotherapy. Herein, paclitaxel (PTX)-loaded poly(lactide-co-glycolide) (PLGA) NBs are prepared as drug carriers with covalently conjugated herceptin (anti-HER2 monoclonal antibody) on the surface to guide the target. The effect of ultrasound on the drug release and targeting of the herceptin-conjugated drug-loaded nanobubbles (PTX-NBs-HER) on the cancerous cells is determined. The use of ultrasound significantly improves the cell targeting capability in vitro, and efficiency of enhanced permeability and retention in vivo. The combination of PTX-NBs-HER and ultrasound facilitates the release of PTX, as well as the uptake and cell apoptosis in vitro. The in vivo application of both PTX-NBs-HER and ultrasound enhances the PTX targeting and accumulation in breast cancers while reducing the transmission and distribution of PTX in healthy organs. The combination of ultrasound with PTX-NBs-HER as contrast agents and drug carriers affords an image-guided drug delivery system for the precise targeted therapy of tumors.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Nanocápsulas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Trastuzumab/administración & dosificación , Animales , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Paclitaxel , Trastuzumab/farmacocinética , Trastuzumab/uso terapéutico , Ondas Ultrasónicas
6.
Biomacromolecules ; 20(1): 401-411, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30485741

RESUMEN

Functionalized nanomaterials with near-infrared (NIR) responsive capacity are quite promising for theranostic treatment of tumors, but formation of NIR responsive nanomaterials with enhanced theranostic ability and excellent biocompatibility is still very challenging. Herein, PEGylated indocyanine green (ICG)-loaded polypyrrole nanoparticles (PPI NPs) were designed and successfully formed through selecting polydopamine as the linkage between each component, demonstrating enhanced NIR responsive theranostic ability against tumor. By combining in vitro cell study with in vivo assay, the formed PPI NPs were proven to be fantastically biocompatible while effectively internalization in HeLa cells and retention in HeLa tumor were demonstrated by in vitro flow cytometry/confocal measurement and in vivo photoacoustic imaging assay. With the guidance of photoacoustic imaging, successful photothermal ablation of tumor was achieved by treatment with PPI NPs plus laser, which was much more effective than the group treated with NPs free of ICG. The combined enhanced photoacoustic and photothermal effect is mainly ascribed to the functionalized polypyrrole nanoparticles, which could accumulate in the tumor site more effectively with a relatively longer retention time taking advantage of the nanomaterial-induced endothelial leakiness phenomenon. All these results demonstrating that this designed PPI NPs possessing enhanced NIR responsive property hold great promise for tumor NIR theranostic applications.


Asunto(s)
Hipertermia Inducida/métodos , Nanopartículas/química , Neoplasias Experimentales/terapia , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Animales , Células HeLa , Humanos , Verde de Indocianina/química , Indoles/química , Rayos Infrarrojos/uso terapéutico , Ratones , Ratones Desnudos , Nanopartículas/efectos adversos , Nanopartículas/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Polietilenglicoles/química , Polímeros/química , Pirroles/química
7.
BMC Cancer ; 14: 800, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25367065

RESUMEN

BACKGROUND: Superparamagnetic poly (lactic-co-glycolic acid) (PLGA)-coated Fe3O4 microcapsules are receiving increased attention as potential diagnostic and therapeutic modalities in the field of oncology. In this study, PLGA-coated Fe3O4 microcapsules were combined with a magnetic resonance imaging-guided high-intensity focused ultrasound (MR-guided HIFU) platform, with the objective of investigating the effects of these composite microcapsules regarding MR-guided HIFU liver cancer surgery in vivo. METHODS: PLGA-coated Fe3O4 microcapsules consisting of a liquid core and a PLGA-Fe3O4 shell were fabricated using a modified double emulsion evaporation method. Their acute biosafety was confirmed in vitro using MDA cells and in vivo using rabbits. To perform MR-guided HIFU surgery, the microcapsules were intravenously injected into a rabbit liver tumor model before MR-guided HIFU. T2-weighted images and MR signal intensity in normal liver parenchyma and tumor tissue were acquired before and after injection, to assess the MR imaging ability of the microcapsules. After MR-guided HIFU ablation tissue temperature mapping, the coagulative volume and histopathology of the tumor tissue were analyzed to investigate the ablation effects of MR-guided HIFUs. RESULTS: Scanning and transmission electron microscopy showed that the microcapsules displayed a spherical morphology and a shell-core structure (mean diameter, 587 nm). The hysteresis curve displayed the typical superparamagnetic properties of the microcapsules, which are critical to their application in MR-guided HIFU surgery. In MR-guided HIFU surgery, these microcapsules functioned as an MRI contrast agent, induced significant hyperthermal enhancement (P < 0.05) and significantly enhanced the volume of coagulative necrosis (P < 0.05). CONCLUSIONS: The administration of PLGA-coated Fe3O4 microcapsules is a potentially synergistic technique regarding the enhancement of MR-guided HIFU cancer surgery.


Asunto(s)
Cápsulas/química , Cápsulas/uso terapéutico , Óxido Ferrosoférrico/química , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Neoplasias Hepáticas/terapia , Polímeros/química , Animales , Cápsulas/administración & dosificación , Modelos Animales de Enfermedad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/ultraestructura , Conejos , Factores de Tiempo
8.
Int J Nanomedicine ; 19: 6499-6513, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38946887

RESUMEN

Purpose: To address the problem of suboptimal reactive oxygen species (ROS) production in Radiation therapy (RT) which was resulted from exacerbated tumor hypoxia and the heterogeneous distribution of radiation sensitizers. Materials and Methods: In this work, a novel nanomedicine, designated as PLGA@IR780-Bi-DTPA (PIBD), was engineered by loading the radiation sensitizer Bi-DTPA and the photothermal agent IR780 onto poly(lactic-co-glycolic acid) (PLGA). This design leverages the tumor-targeting ability of IR780 to ensure selective accumulation of the nanoparticles in tumor cells, particularly within the mitochondria. The effect of the photothermal therapy-enhanced radiation therapy was also examined to assess the alleviation of hypoxia and the enhancement of radiation sensitivity. Results: The PIBD nanoparticles exhibited strong capacity in mitochondrial targeting and selective tumor accumulation. Upon activation by 808 nm laser irradiation, the nanoparticles effectively alleviated local hypoxia by photothermal effect enhanced blood supplying to improve oxygen content, thereby enhancing the ROS production for effective RT. Comparative studies revealed that PIBD-induced RT significantly outperformed conventional RT in treating hypoxic tumors. Conclusion: This design of tumor-targeting photothermal therapy-enhanced radiation therapy nanomedicine would advance the development of targeted drug delivery system for effective RT regardless of hypoxic microenvironment.


Asunto(s)
Nanopartículas , Terapia Fototérmica , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Especies Reactivas de Oxígeno , Animales , Terapia Fototérmica/métodos , Especies Reactivas de Oxígeno/metabolismo , Nanopartículas/química , Línea Celular Tumoral , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratones , Indoles/farmacología , Indoles/química , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias/radioterapia , Neoplasias/terapia , Neoplasias/metabolismo , Nanomedicina
9.
Biomaterials ; 308: 122570, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636133

RESUMEN

Metallic biomaterials activate tumor ferroptosis by increasing oxidative stress, but their efficacy is severely limited in tumor microenvironment. Although interferon gamma (IFN-γ) can promote tumor ferroptosis sensitivity by inhibiting the antioxidant system and promoting lipid accumulation, this effect limited by the lack of IFN-γ accumulation in tumors. Herein, we report a near-infrared (NIR)-responsive HCuS nanocomposite (HCuS-PE@TSL-tlyp-1) that can stimulate immunogenic cell death (ICD)-mediated IFN-γ secretion through exogenous oxidative stress, thereby achieving cascaded ferrotherapy by mutually reinforcing ferroptosis and systemic immunity. Upon laser irradiation, the dissolution of the thermal coating, and the introduction of Cu ions and piperazine-erastin (PE) simultaneously induce oxidative stress by reactive oxygen species (ROS)/lipid peroxide (LPO) accumulation and deplete cystine-glutamate transporter (xCT)/GSH. The onset of oxidative stress-mediated ferroptosis is thus achieved, and ICD is triggered, significantly promoting cytotoxic T-cell (CTL) infiltration for IFN-γ secretion. Furthermore, IFN-γ induces immunogenic tumor ferroptosis by inhibiting xCT-antioxidant pathways and enhancing the ACSL4-fatty acid recruitment pathway, which further promotes sensitivity to ferroptosis in cells. These HCuS nanocomposites combined with aPD-L1 effectively in inhibiting tumor metastasis and recurrence. Importantly, these cascade ferrotherapy results broadens the application of HCuS biomaterials.


Asunto(s)
Cobre , Ferroptosis , Interferón gamma , Liposomas , Ferroptosis/efectos de los fármacos , Animales , Cobre/química , Cobre/farmacología , Interferón gamma/metabolismo , Ratones , Liposomas/química , Nanocompuestos/química , Línea Celular Tumoral , Muerte Celular Inmunogénica/efectos de los fármacos , Rayos Infrarrojos , Humanos , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
10.
J Microencapsul ; 29(5): 437-44, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22299595

RESUMEN

The purpose of this study was to develop a sonosensitizer-loaded multi-functional ultrasound (US) contrast agent for both tumour therapy and imaging. The hematoporphyrin (HP)-encapsulated poly(lactic-co-glycolic acid) microbubbles (HP-PLGA-MBs) were prepared and filled with perfluorocarbon gases. The enhancement of US imaging and its sonodynamically induced anti-tumour effect were evaluated by both in vitro and in vivo experiments. The HP-PLGA-MBs have a narrow size distribution and smooth surface with a mean diameter of 702.6 ± 56.8 nm and HP encapsulation efficiency of 63.50 ± 1.26% and drug-loading efficiency of 2.15 ± 0.13%. The HP-PLGA-MBs could well enhance the ultrasound imaging both in vitro and in vivo. A significant anti-tumour effect was obtained by HP-PLGA-MBs mediated sonodynamic therapy. The tumour growth rate and the tumour proliferation index were the lowest in the HP-PLGA-MBs plus sonication group. And the tumour cell apoptotic index was the biggest in the HP-PLGA-MBs plus sonication group. In conclusion, a sonosensitizer-loaded multi-functional contrast agent was constructed and the feasibility was demonstrated, which might provide a novel strategy for tumour imaging and therapy.


Asunto(s)
Carcinoma Hepatocelular/terapia , Medios de Contraste/uso terapéutico , Hematoporfirinas/uso terapéutico , Neoplasias Hepáticas/terapia , Ultrasonido/instrumentación , Animales , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Medios de Contraste/administración & dosificación , Diseño de Equipo , Hematoporfirinas/administración & dosificación , Ácido Láctico/química , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/patología , Ratones , Microburbujas/uso terapéutico , Tamaño de la Partícula , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Terapia por Ultrasonido/instrumentación
11.
Nanoscale ; 14(33): 12069-12076, 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-35947015

RESUMEN

Near-infrared (NIR) laser triggered theranostic platforms are increasingly used in clinical nanomedicine applications. In this work, a core-shell composite consisting of polypyrrole (PPy) coated copper sulfide (CuS) nanospheres with high photothermal efficiency and good photostability has been fabricated via a facile interfacial polymerization. The PPy@CuS nanohybrid had a hydrodynamic diameter of 58.5 nm with a CuS core and PPy shell and exhibited strong optical absorption and photon-to-heat conversion in the NIR region, leading to a sufficient photohyperthermic effect under irradiation with a 808 nm continuous wave laser. In vivo studies showed that the Ppy@CuS nanohybrids produced significant photoacoustic signals and exhibited remarkable photothermal therapeutic efficacy. Furthermore, the core-shell composites exhibited improved temperature elevation and photostability. The temperature-induced changes can be detected and monitored using photoacoustic imaging, thus allowing the control of the thermal dose while minimizing photothermal damage to surrounding healthy tissues. In summary, this study demonstrates that this novel platform could potentially be used for photoacoustic image-guided photothermal therapy and real-time temperature monitoring in cancer theranostics.


Asunto(s)
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Cobre , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Técnicas Fotoacústicas/métodos , Fototerapia , Terapia Fototérmica , Polímeros , Pirroles , Temperatura , Nanomedicina Teranóstica/métodos
12.
Curr Med Chem ; 29(8): 1352-1368, 2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-34137360

RESUMEN

BACKGROUND: Advances in nanotechnology have enabled the combination of disease diagnosis and therapy into a single nano package that has tremendous potential for the development of new theranostic strategies. The variety of polymer-based materials has grown exponentially over the past several decades. Such materials have great potential as carriers in disease detection imaging and image monitoring and in systems for the precise delivery of drugs to specific target sites. OBJECTIVE: In the present article, we review recent key developments in the synthesis of polymer- based materials for various medical applications and their clinical trials. CONCLUSION: There is a growing range of multi-faceted, polymer-based materials with various functions. These functions include carriers for image contrast agents, drug delivery systems, and real-time image-guided systems for noninvasive or minimally invasive therapeutic procedures for cancer therapy.


Asunto(s)
Neoplasias , Polímeros , Portadores de Fármacos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanotecnología , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Polímeros/uso terapéutico , Nanomedicina Teranóstica
13.
Drug Deliv ; 29(1): 3111-3122, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36131565

RESUMEN

Mitochondria play an important role in regulating tumor cell death and metabolism so that they can be potential therapeutic targets. Sonodynamic therapy (SDT) represents an attractive antitumor method that induces apoptosis by producing highly toxic reactive oxygen species (ROS). Mitochondria-targeting SDT can cause oxidative damage and improve the efficiency of tumor therapy. However, due to the nonselective distribution of nanosystems and the anti-apoptotic mechanism of cancer cells, the therapeutic effect of SDT is not ideal. Therefore, we proposed a novel mitochondria-targeting nanosystem ('Mito-Bomb') for ferroptosis-boosted SDT. Sonosensitizer IR780 and ferroptosis activator RSL-3 were both encapsulated in biocompatible poly(lactic-co-glycolic acid) (PLGA) nanoparticles to form 'Mito-Bomb' (named IRP NPs). IR780 in this nanosystem was used to mediate mitochondria-targeting SDT. RSL-3 inhibited the activity of GPX4 in the antioxidant system to induce ferroptosis of tumor cells, which could rewire tumor metabolism and make tumor cells extremely sensitive to SDT-induced apoptosis. Notably, we also found that RSL-3 can inhibit hypoxia inducible factor-1α (HIF-1α) and induce ROS production to improve the efficacy of SDT to synergistically antitumor. RSL-3 was applied as a 'One-Stone-Three-Birds' agent for cooperatively enhanced SDT against triple-negative breast cancer. This study presented the first example of RSL-3 boosting mitochondria-targeting SDT as a ferroptosis activator. The 'Mito-Bomb' biocompatible nanosystem was expected to become an innovative tumor treatment method and clinical transformation.


Asunto(s)
Bombas (Dispositivos Explosivos) , Ferroptosis , Terapia por Ultrasonido , Antioxidantes/metabolismo , Línea Celular Tumoral , Mitocondrias , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Terapia por Ultrasonido/métodos
14.
Drug Deliv ; 29(1): 2177-2189, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35815688

RESUMEN

We fabricated an ultrasound activated 'nanobomb' as a noninvasive and targeted physical therapeutic strategy for sonodynamic therapy and priming cancer immunotherapy. This 'nanobomb' was rationally designed via the encapsulation of indocyanine green (ICG) and perfluoropentane (PFP) into cRGD peptide-functionalized nano-liposome. The resulting Lip-ICG-PFP-cRGD nanoparticle linked with cRGD peptide could actively targeted ID8 and TC-1 cells and elicits ROS-mediated apoptosis after triggered by low-intensity focused ultrasound (LIFU). Moreover, the phase change of PFP (from droplets to microbubbles) under LIFU irradiation can produce a large number of microbubbles, which act as intra-tumoral bomber and can detonate explode tumor cells by acoustic cavitation effect. Instant necrosis of tumor cells further induces the release of biologically active damage-associated molecular patterns (DAMPs) to facilitate antitumor immunity. More important, the 'nanobomb' in combination with anti-PD-1checkpoint blockade therapy can significantly improve the antitumor efficacy in a subcutaneous model. In addition, the liposomes may also be used as an imaging probe for ultrasound (US) imaging after being irradiated with LIFU. In summary, the US imaging-guided, LIFU activated ROS production and explosion 'nanobomb' might significantly improve the antitumor efficacy and overcome drug resistance through combination of SDT and immunotherapy, we believe that this is a promising approach for targeted therapy of solid tumor including ovarian cancer.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Acústica , Línea Celular Tumoral , Femenino , Humanos , Verde de Indocianina/química , Liposomas/química , Microburbujas , Nanopartículas/química , Neoplasias Ováricas/tratamiento farmacológico , Especies Reactivas de Oxígeno , Ultrasonografía
15.
Int J Nanomedicine ; 17: 4547-4565, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199475

RESUMEN

Background: Sonodynamic therapy (SDT) has been regarded as a novel therapeutic modality for killing tumors. However, the hypoxic tumor microenvironment, especially deep-seated tumors distant from blood vessels, severely restricts therapeutic efficacy due to the oxygen-dependent manner of SDT. Methods: Herein, we report a novel ultrasonic cavitation effect-based therapeutic modality that is able to facilitate the hypoxia-tolerant SDT for inducing hypoxic tumor death. A tLyP-1 functionalized liposomes is fabricated, composed of hematoporphyrin monomethyl ether gadolinium as the sonosentizer and perfluoropentane (PFP) as the acoustic environment regulator. Moreover, the tLyP-1 functioned liposomes could achieve active tumor homing and effective deep-penetrating into hypoxic tumors. Upon low intensity focused ultrasound (LIFU) irradiation, the acoustic droplet vaporization effect of PFP induced fast liquid-to-gas transition and quick bubbles explosion to generate hydroxyl radicals, efficiently promoting cell death in both normoxic and hypoxic microenvironment (acting as deep-penetration nanobomb, DPNB). Results: The loading of PFP is proved to significantly enhance the therapeutic efficacy of hypoxic tumors. In particular, these DPNB can also act as ultrasound, photoacoustic, magnetic resonance, and near-infrared fluorescence tetramodal imaging agents for guiding the therapeutic process. Conclusion: This study is the first report involving that liquid-to-gas transition based SDT has the potential to combat hypoxic tumors.


Asunto(s)
Neoplasias , Terapia por Ultrasonido , Línea Celular Tumoral , Gadolinio , Humanos , Hipoxia/diagnóstico por imagen , Hipoxia/terapia , Liposomas , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Neoplasias/terapia , Oxígeno , Microambiente Tumoral , Terapia por Ultrasonido/métodos
16.
ACS Appl Mater Interfaces ; 13(3): 3605-3621, 2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33449625

RESUMEN

Breast cancer is a major threat to health and lives of females. Biomimetic nanotechnology brought brighter hope for early diagnosis and treatment of breast cancer. Here, we proposed a platelet (PLT) membrane-derived strategy for enhanced photoacoustic (PA)/ultrasonic (US)/fluorescence (FL) multimodal imaging and augmented synergistic photothermal/chemotherapeutic efficacy in tumor cells. A PA imaging contrast and photothermal agent, nanocarbons (CNs), a chemotherapeutic and FL material, doxorubicin (DOX), and perfluoropentane (PFP) were coencapsulated into the poly(lactic-co-glycolic) acid (PLGA) skeletons. Then, the PLT membranes were coated onto the PLGA NPs, which were named as "nanoplatelets" (DOX-PFP-CNs@PLGA/PM NPs). The "nanoplatelets", which conserved the structural advantages and inherent properties of PLTs, could not only escape from phagocytosis of macrophages but also actively targeted tumor cells by the way of antigen-antibody interactions between P-selectin on the PM and CD44 receptors of the tumor cells. With CNs and DOX loaded in, these "nanoplatelets" could serve as an excellent contrast agent for PA/FL imaging. Under laser irradiation, the "nanoplatelets" could turn light energy into heat energy. The laser-triggered photothermal effect, on the one hand, could ablate the tumor cells immediately, and on the other hand, could initiate the optical droplet vaporization of PFP, which subsequently enhanced US imaging and promoted the discharge of encapsulated DOX from the "nanoplatelets" for remarkably strengthening photothermal therapeutic power in turn. In this work, as compared with the bare drug-loaded nanoparticles, the "nanoplatelets" exhibited much more accumulation in the tumor cells, demonstrating superior multimodal imaging capability and preferable synergistic therapeutic performance. In conclusion, the "nanoplatelets" could serve as contrast agents for US imaging and PA imaging to guide the therapy. What is more, the bioinspired PLT-derived, targeted, and nontoxic "nanoplatelets", which were exploited for multimodal PA/US/FL imaging-guided synergistic photothermal/chemo therapy, will be of great value to breast cancer theranostics in the days to come.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/uso terapéutico , Materiales Biomiméticos/química , Biomimética , Neoplasias de la Mama/patología , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Imagen Óptica/métodos , Técnicas Fotoacústicas/métodos , Terapia Fototérmica , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Nanomedicina Teranóstica/métodos
17.
Biomed Res Int ; 2020: 6863231, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33015175

RESUMEN

Gold nanorods exhibit a wide variety of applications such as tumor molecular imaging and photothermal therapy (PTT) due to their tunable optical properties. Several studies have demonstrated that the combination of other therapeutic strategies may improve PTT efficiency. A method called optical droplet vaporization (ODV) was considered as another noninvasive imaging and therapy strategy. Via the ODV method, superheated perfluorocarbon droplets can be vaporized to a gas phase for enhancing ultrasound imaging; meanwhile, this violent process can cause damage to cells and tissue. In addition, active targeting through the functionalization with targeting ligands can effectively increase nanoprobe accumulation in the tumor area, improving the sensitivity and specificity of imaging and therapy. Our study prepared a nanoparticle loaded with gold nanorods and perfluorinated hexane and conjugated to a monoclonal antibody (MAGE-1 antibody) to melanoma-associated antigens (MAGE) targeting melanoma, investigated the synergistic effect of PTT/ODV therapy, and monitored the therapeutic effect using ultrasound. The prepared MAGE-Au-PFH-NPs achieved complete eradication of tumors. Meanwhile, the MAGE-Au-PFH-NPs also possess significant ultrasound imaging signal enhancement, which shows the potential for imaging-guided tumor therapy in the future.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Oro/química , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/terapia , Nanopartículas del Metal/química , Fototerapia , Neoplasias Cutáneas/terapia , Ultrasonografía , Animales , Materiales Biocompatibles , Proteínas de Choque Térmico/metabolismo , Hipertermia Inducida , Masculino , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Óptica , Neoplasias Cutáneas/diagnóstico por imagen , Pruebas de Toxicidad
18.
Biomater Sci ; 8(3): 858-870, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31808470

RESUMEN

Hypoxic resistance, photosensitizer toxicity, and target deficiency are major challenges strongly inhibiting the efficacy of clinical photodynamic therapy (PDT) in tumor treatment. To overcome these challenges, we synthesized IR780 and catalase co-loaded liposomes to form a tumor-targeted bio-nanoreactor (LIP-IR-CAT). The efficient strategy can solve the physicochemical problems including strong hydrophobicity, poor light stability, poor tolerance, and high toxicity in vivo of IR780 as a photosensitizer and promote the clinical application of IR780. Taking advantage of the high catalytic efficiency of catalase when it meets hydrogen peroxide (H2O2), continuous oxygen can be generated due to the abnormally elevated level of H2O2 within the tumor, thus remarkably promoting tumor oxygenation. With the conjunction of photosensitivity and specific mitochondria-targeting ability of IR780, the intratumoral reactive oxygen species (ROS) are strongly enhanced, and adenosine triphosphate (ATP) is reduced under near-infrared (NIR) laser irradiation. Following a single-dose intravenous injection of LIP-IR-CAT, tumor hypoxia can be seriously attenuated, at the same time creating an opportunity to enhance the efficacy of PDT on the tumor. Our in vivo data show that the nanoreactor LIP-IR-CAT, in combination with just two short time NIR laser irradiation sessions, can effectively inhibit the growth of solid tumors without systemic toxicity.


Asunto(s)
Peróxido de Hidrógeno/administración & dosificación , Hipoxia/terapia , Neoplasias/terapia , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Catalasa/administración & dosificación , Catalasa/química , Línea Celular Tumoral , Femenino , Humanos , Peróxido de Hidrógeno/química , Hipoxia/metabolismo , Indoles/administración & dosificación , Indoles/química , Rayos Infrarrojos , Liposomas/administración & dosificación , Liposomas/química , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Neoplasias/metabolismo , Oxígeno/análisis , Oxígeno/metabolismo , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Especies Reactivas de Oxígeno/metabolismo
19.
ACS Appl Mater Interfaces ; 12(43): 48420-48431, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33073973

RESUMEN

Photothermal therapy (PTT) as a noninvasive and effective thermal therapeutic approach has attracted tremendously increasing interest because it can effectively eliminate the primary tumor and generate tumor-associated antigens, which could elicit antitumor immune responses. Herein, we report on the rational design and fabrication of copper sulfide (CuS)-based nanoplatform for cancer photothermal immunotherapy. The as-prepared core-shell CuS@mSiO2-PFP-PEG (CPPs) nanocomposites possess high biocompatibility, photoacoustic (PA)/ultrasound (US) imaging, and strong PTT effect upon 808 nm laser irradiation, indicating that the nanocomposites have a promising application in diagnosis and treatment of breast cancer with molecular classification. Importantly, we also elucidated that the CPP-triggered PTT in combination with anti-PD-1 checkpoint blockade therapy can not only obliterate primary tumor but also inhibit metastatic tumor in tumor-bearing mice. We believe that the CPPs have a good probability to serve as a useful nanoplatform for PTT, and this approach may provide a promising strategy for tumor-therapeutic modality with immunotherapy.


Asunto(s)
Inmunoterapia , Luz , Nanocompuestos/química , Terapia Fototérmica , Animales , Supervivencia Celular/efectos de los fármacos , Cobre/química , Fluorocarburos/química , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Técnicas Fotoacústicas , Polietilenglicoles/química , Dióxido de Silicio/química , Sulfuros/química , Propiedades de Superficie , Células Tumorales Cultivadas , Ultrasonografía
20.
Int J Nanomedicine ; 15: 1759-1770, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32214809

RESUMEN

BACKGROUND: There have been many recent reports of molecular probes for thrombi but with unsatisfactory in vivo targeting effects, which could be related to the blood flow velocity in vivo. Therefore, it is worth explaining the relationship between the targeting effect and the blood flow velocity. METHODS AND MATERIALS: In this study, we constructed a platelet-targeting nanoparticle (NP) based on EWVDV for targeting P-selectin combined with the phase transition material perfluorohexane and India ink to achieve the multimodal imaging of thrombi. We studied the targeting effect of the NPs for rabbit blood thrombi under different flow velocities simulating blood flow velocities in vivo. RESULTS: The results show the successful fabrication of NPs with the ability to undergo a phase transition via low-intensity focused ultrasound irradiation to achieve ultrasound imaging and with a high binding affinity for activated platelets. In vitro, low flow velocities (20 cm/s) hardly affected the targeting effect of the NPs, while moderate flow velocities (40 cm/s) reduced the number of NPs that target thrombi by 52.6% comparing to static fluid (0 cm/s). High flow velocities (60 cm/s) greatly reduced the targeting effect of the NPs by 83.5%. CONCLUSION: These results can serve as a reference for the design of NPs targeting thrombi at different sites and in different blood vessel types according to the blood flow velocity, thereby establishing a foundation for in vivo experiments.


Asunto(s)
Plaquetas/efectos de los fármacos , Imagen Multimodal/métodos , Nanopartículas/administración & dosificación , Trombosis/diagnóstico por imagen , Animales , Velocidad del Flujo Sanguíneo , Carbono/química , Fluorocarburos/química , Nanopartículas/química , Selectina-P/metabolismo , Activación Plaquetaria/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Conejos , Trombosis/sangre , Ultrasonografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA