Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Med Sci Monit ; 28: e934282, 2022 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-35124688

RESUMEN

BACKGROUND This study compared the effects of peritoneal dialysis and hemodialysis on cognitive dysfunction and health-related quality of life (HRQOL) in end-stage renal disease (ESRD) patients and analyzed other potential influencing factors. MATERIAL AND METHODS A total of 265 patients who received dialysis at our hospital were included and divided into the hemodialysis group (n=115) and the peritoneal dialysis group (n=150). The cognitive performance was assessed by the Beijing version of the Montreal Cognitive Assessment. The Kidney Disease Quality of 36-Item Short Form Survey and a kidney disease-related quality of life assessment were used for evaluating HRQOL. Univariate and multivariate linear regression analyses were used to explore the effects of dialysis on cognitive dysfunction and HRQOL. RESULTS As compared with the hemodialysis group, the peritoneal dialysis group had lower scores on the Montreal Cognitive Assessment (ß=-8.35, 95% CI: -9.85 to -6.86), 36-Item Short Form Survey (ß=-10.20, 95% CI: -11.94 to -8.45), and kidney disease-related quality of life assessment (ß=-8.67, 95% CI: -10.10 to -7.23). After adjustment for sex, age, BMI, marital status, educational level, income level, presence of diabetes, duration of kidney disease, duration of dialysis, and dialysis frequency, the results were consistent with that of the crude model. CONCLUSIONS In the present study, patients receiving peritoneal dialysis had worse cognitive dysfunction and worse HRQOL compared to patients receiving hemodialysis, which might lead to poorer outcomes of ESRD patients. The related factors affecting cognitive dysfunction and HRQOL were also explored, which could help clinicians to determine the optimal treatment for ESRD patients.


Asunto(s)
Disfunción Cognitiva/epidemiología , Encuestas Epidemiológicas/métodos , Fallo Renal Crónico/epidemiología , Fallo Renal Crónico/terapia , Diálisis Peritoneal/métodos , Calidad de Vida/psicología , Diálisis Renal/métodos , Beijing/epidemiología , Disfunción Cognitiva/psicología , Estudios Transversales , Femenino , Humanos , Fallo Renal Crónico/psicología , Masculino , Persona de Mediana Edad , Diálisis Peritoneal/psicología , Diálisis Renal/psicología
2.
Handb Exp Pharmacol ; 273: 351-364, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33454856

RESUMEN

For a chemotherapeutic agent to be effective, it must conquer the presence of blood-brain barrier (BBB), which limits the penetration of drugs into the brain. Tumours in the brain compromise the integrity of BBB and result in a highly heterogeneous vasculature, known as blood-brain tumour barrier (BBTB). In this chapter, we firstly highlight the cellular and molecular characteristics of the BBB and BBTB as well as the challenges aroused by BBB/BBTB for drug delivery. Secondly, we discuss the current strategies overcoming the challenges in invasive and non-invasive manners. Finally, we highlight the emerging strategy using focused ultrasound (FUS) with systemic microbubbles to transiently and reversibly enhance the permeability of these barriers for drug delivery.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Encéfalo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Sistemas de Liberación de Medicamentos , Humanos , Microburbujas
3.
Sensors (Basel) ; 22(10)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35632027

RESUMEN

Carton detection is an important technique in the automatic logistics system and can be applied to many applications such as the stacking and unstacking of cartons and the unloading of cartons in the containers. However, there is no public large-scale carton dataset for the research community to train and evaluate the carton detection models up to now, which hinders the development of carton detection. In this article, we present a large-scale carton dataset named Stacked Carton Dataset (SCD) with the goal of advancing the state-of-the-art in carton detection. Images were collected from the Internet and several warehouses, and objects were labeled for precise localization using instance mask annotation. There were a total of 250,000 instance masks from 16,136 images. Naturally, a suite of benchmarks was established with several popular detectors and instance segmentation models. In addition, we designed a carton detector based on RetinaNet by embedding our proposed Offset Prediction between the Classification and Localization module (OPCL) and the Boundary Guided Supervision module (BGS). OPCL alleviates the imbalance problem between classification and localization quality, which boosts AP by 3.1∼4.7% on SCD at the model level, while BGS guides the detector to pay more attention to the boundary information of cartons and decouple repeated carton textures at the task level. To demonstrate the generalization of OPCL for other datasets, we conducted extensive experiments on MS COCO and PASCAL VOC. The improvements in AP on MS COCO and PASCAL VOC were 1.8∼2.2% and 3.4∼4.3%, respectively.


Asunto(s)
Compuestos Orgánicos Volátiles
4.
Curr Pain Headache Rep ; 24(11): 68, 2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-32990832

RESUMEN

PURPOSE OF REVIEW: This review provides an update on chronic cluster headache (CH) focusing on clinical features, pathophysiology, and management as well as comparisons between Eastern and Western populations. RECENT FINDINGS: Chronic CH in Eastern populations was relatively rare, compared to that in Western populations. Lacrimation and/or conjunctival injection is the most frequently reported cranial autonomic symptom, and visual aura is predominant in chronic CH patients. Neuroimaging evidence in both ethnic groups suggests that CH pathophysiology involves the hypothalamus and pain-modulatory areas, with dynamic alternations between CH episodes. Recent evidence indicates that midbrain dopaminergic systems may participate in CH chronicity. Noteworthy advances have emerged in neuromodulatory therapies for chronic CH, but treatment with calcitonin gene-related peptide (CGRP) monoclonal antibodies has been unsuccessful. Recent evidence shows divergence of chronic CH between Eastern and Western populations. Neuromodulatory therapies but not CGRP inhibition is effective in this intractable patient group.


Asunto(s)
Cefalalgia Histamínica/epidemiología , Cefalalgia Histamínica/fisiopatología , Trastornos de Cefalalgia/epidemiología , Trastornos de Cefalalgia/fisiopatología , Cefalalgia Histamínica/terapia , Trastornos de Cefalalgia/terapia , Humanos
5.
Int J Hyperthermia ; 35(1): 599-611, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30295119

RESUMEN

PURPOSE: Ultrasound contrast agent microbubbles were combined with magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS) as a means to achieve mild hyperthermia at reduced power levels. METHODS: MRgFUS hyperthermia (42°C for 20 min) was evaluated in rabbit thigh muscle or Vx2 tumors using infusions of microbubbles (Definity, 20 µL/kg) or saline (sham) administered over 5 min. The impact of treatments on drug uptake was assessed with liposomal doxorubicin (Caelyx, 2.5 mg/kg). Applied power levels before and after the injection of microbubbles or saline were compared, and drug uptake was evaluated with fluorometry of tissues harvested 24 hr post-treatment. RESULTS: MRgFUS hyperthermia in muscle and tumors resulted in accurate temperature control (mean =42.0°C, root mean square error (RMSE) = 0.3°C). The power dropped significantly following the injection of microbubbles in muscle and tumors compared to exposures without microbubbles (-21.9% ± 12.5% vs -5.9% ± 7.8%, p = .009 in muscle; -33.8% ± 9.9% vs -3.0% ± 7.2%, p < .001 in tumors). Cavitation monitoring indicated emission of subharmonic, ultraharmonic, and elevated levels of fourth to sixth harmonic frequencies following microbubble injection. The drug delivery was elevated significantly in muscle with the use of microbubble-assisted relative to conventional heating (0.5 ± 0.5 ng/mg vs 0.20 ± 0.04 ng/mg, p = .05), whereas in tumors similar levels were found (11 ± 3 ng/mg vs 16 ± 4 ng/mg, p = .13). CONCLUSIONS: The finding that microbubbles reduce the applied power requirements for hyperthermia has considerable clinical implications. The elevated levels of drug found in muscle but not tumor tissue suggest a complex interplay between the heating effects of microbubbles with those of enhanced permeabilization and possible vascular damage.


Asunto(s)
Fiebre/diagnóstico por imagen , Fiebre/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Imagen por Resonancia Magnética/métodos , Microburbujas/uso terapéutico , Animales , Fiebre/patología , Humanos , Masculino , Conejos
6.
Nanotheranostics ; 6(4): 376-387, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795341

RESUMEN

In sonodynamic therapy, cellular toxicity from sonosensitizer drugs, such as 5-aminolevulinic acid hydrochloride (5-ALA), may be triggered with focused ultrasound through the production of reactive oxygen species (ROS). Here we show that by increasing local oxygen during treatment, using oxygen-loaded perfluorocarbon nanodroplets (250 +/- 8 nm), we can increase the damage induced by 5-ALA, and monitor the severity by recording acoustic emissions in the brain. To achieve this, we sonicated the right striatum of 16 healthy rats after an intravenous dose of 5-ALA (200 mg/kg), followed by saline, nanodroplets, or oxygen-loaded nanodroplets. We assessed haemorrhage, edema and cell apoptosis immediately following, 24 hr, and 48 hr after focused ultrasound treatment. The localized volume of damaged tissue was significantly enhanced by the presence of oxygen-loaded nanodroplets, compared to ultrasound with unloaded nanodroplets (3-fold increase), and ultrasound alone (40-fold increase). Sonicating 1 hr following 5-ALA injection was found to be more potent than 2 hr following 5-ALA injection (2-fold increase), and the severity of tissue damage corresponded to the acoustic emissions from droplet vaporization. Enhancing the local damage from 5-ALA with monitored cavitation activity and additional oxygen could have significant implications in the treatment of atherosclerosis and non-invasive ablative surgeries.


Asunto(s)
Lesiones Encefálicas , Oxígeno , Ácido Aminolevulínico , Animales , Encéfalo , Monitoreo Fisiológico , Ratas , Triptófano Oxigenasa , Ultrasonografía
7.
Magn Reson Med ; 65(3): 802-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20941741

RESUMEN

Focused ultrasound combined with an intravascular ultrasound contrast agent can induce transient disruption of the blood-brain barrier, and the blood-brain barrier disruption can be detected by contrast-enhanced MRI. There is, however, no study investigating the ability of various MR methods to detect focused ultrasound-induced blood-brain barrier disruption within minimal hemorrhage. Sonication was applied to 15 rat brains with four different doses of ultrasound contrast agent (0, 10, 30, or 50 µL/kg), and contrast-enhanced T1-weighted spin echo, gradient echo images, and longitudinal relaxation rate mapping along with effective transverse relaxation time-weighted and susceptibility-weighted images were acquired. Volume-of-interest-based and threshold-based analyses were performed to quantify the contrast enhancement, which was then correlated with the ultrasound contrast agent dose and with the amount of Evans blue extravasation. Both effective transverse relaxation time-weighted and susceptibility-weighted images did not detect histology-proved intracranial hemorrhage at 10 µL/kg, but MRI failed to detect mild intracranial hemorrhage at 30 µL/kg. All tested sequences showed detectable contrast enhancement increasing with ultrasound contrast agent dose. In correlating with Evans blue extravasation, the gradient echo sequence was slightly better than the spin echo sequence and was comparable to longitudinal relaxation rate mapping. In conclusion, both gradient echo and spin echo sequences were all reliable in indicating the degree of focused ultrasound-induced blood-brain barrier disruption within minimal hemorrhage.


Asunto(s)
Barrera Hematoencefálica/anatomía & histología , Barrera Hematoencefálica/efectos de la radiación , Gadolinio DTPA , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Sonicación/métodos , Animales , Hemorragia Cerebral/etiología , Hemorragia Cerebral/prevención & control , Medios de Contraste , Masculino , Ratas , Ratas Wistar , Sonicación/efectos adversos
8.
J Control Release ; 332: 30-39, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33600879

RESUMEN

Focused ultrasound (FUS) offers an attractive tool for non-invasive neuromodulation, addressing a clinical need to develop more minimally invasive approaches that are safer, more tolerable and versatile. In combination with a cavitation agent, the effects of ultrasound can be amplified and localized for therapy. Using c-Fos expression mapping, we show how ultrasound-sensitive nanodroplets can be used to induce either neurosuppression or neurostimulation, without disrupting the blood-brain barrier in rats. By repurposing a commercial ultrasound contrast agent, Definity, lipid-shell decafluorobutane-core nanodroplets of 212.5 ± 2.0 nm were fabricated and loaded with or without pentobarbital. FUS was delivered with an atlas-based targeting system at 1.66 MHz to the motor cortex of rats, using a feedback-controller to detect successful nanodroplet vaporization and drug release. Neuromodulation was quantified through changes in sensorimotor function and c-Fos expression. Following FUS-triggered delivery, sham nanodroplets induced a 22.6 ± 21% increase in local c-Fos expression, whereas pentobarbital-loaded nanodroplets induced a 21.7 ± 13% decrease (n = 6). Nanodroplets, combined with FUS, offer an adaptable tool for neuromodulation, through local delivery of small molecule anesthetics or targeted mechanical effects.


Asunto(s)
Barrera Hematoencefálica , Medios de Contraste , Animales , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Pentobarbital , Ratas , Ultrasonografía
9.
J Magn Reson Imaging ; 31(6): 1323-30, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512883

RESUMEN

PURPOSE: To optimize the timing of contrast-enhanced magnetic resonance imaging (MRI) that best indicates blood-brain barrier (BBB) disruption induced by focused ultrasound (FUS) along with an ultrasound contrast agent (UCA) and to verify that the contrast-enhanced spin-echo MRI sequence can indicate the degree and location of BBB disruption in the presence of hemorrhage better than a gradient-echo sequence. MATERIALS AND METHODS: Sonication was applied to 12 rat brains with four different doses of UCA to cause variable degrees of hemorrhage. Two imaging sequences were performed to acquire T1-weighted (T1W) images at two time-points after the administration of a T1-shortening contrast agent. The contrast enhancement at the sonicated regions was quantified and correlated against Evans blue (EB) staining. RESULTS: The spin-echo T1W images at 10 minutes post-contrast enhancement showed the best correlation with EB staining in both quantity of EB extravasation (r = 0.812; P < 0.01) and spatial distribution (r = 0.528, P < 0.01). This capability was more robust than the gradient-echo sequence. CONCLUSION: Our results suggest that contrast-enhanced T1W spin-echo sequence acquired in the early phase post-contrast enhancement should be considered to monitor the degree and location of BBB disruption under the possibility of hemorrhage induced by FUS.


Asunto(s)
Barrera Hematoencefálica/patología , Hemorragia/patología , Imagen por Resonancia Magnética/métodos , Animales , Encéfalo/patología , Medios de Contraste/farmacología , Azul de Evans/farmacología , Procesamiento de Imagen Asistido por Computador , Masculino , Ratas , Ratas Wistar , Sonicación , Factores de Tiempo , Ultrasonografía/métodos
10.
Pharmaceutics ; 13(1)2020 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-33374205

RESUMEN

The presence of blood-brain barrier (BBB) and/or blood-brain-tumor barriers (BBTB) is one of the main obstacles to effectively deliver therapeutics to our central nervous system (CNS); hence, the outcomes following treatment of malignant brain tumors remain unsatisfactory. Although some approaches regarding BBB disruption or drug modifications have been explored, none of them reach the criteria of success. Convention-enhanced delivery (CED) directly infuses drugs to the brain tumor and surrounding tumor infiltrating area over a long period of time using special catheters. Focused ultrasound (FUS) now provides a non-invasive method to achieve this goal via combining with systemically circulating microbubbles to locally enhance the vascular permeability. In this review, different approaches of delivering therapeutic agents to the brain tumors will be discussed as well as the characterization of BBB and BBTB. We also highlight the mechanism of FUS-induced BBB modulation and the current progress of this technology in both pre-clinical and clinical studies.

11.
Sci Adv ; 6(36)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917589

RESUMEN

Thermosensitive liposomes represent an important paradigm in oncology, where hyperthermia-mediated release coupled with thermal bioeffects enhance the effectiveness of chemotherapy. Their widespread clinical adoption hinges upon performing controlled targeted hyperthermia, and a leading candidate to achieve this is temperature-based magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS). However, the current approach to hyperthermia involves exposures lasting tens of minutes to hours, which is not possible to achieve in many circumstances because of blood vessel cooling and respiratory motion. Here, we investigate a novel approach to overcome these limitations: to use fractionated ultrashort (~30 s) thermal exposures (~41° to 45°C) to release doxorubicin from a thermosensitive liposome. This is first demonstrated in a dorsal chamber tumor model using two-photon microscopy. Thermal exposures were then conducted with a rabbit tumor model using a custom MRgFUS system incorporating temperature feedback control. Drug release was confirmed, and longitudinal experiments demonstrated profoundly enhanced tumor growth inhibition and survival.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias , Animales , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Liposomas , Imagen por Resonancia Magnética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Conejos
12.
Sci Rep ; 9(1): 10465, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320671

RESUMEN

Glioblastoma multiforme (GBM) continues to have a dismal prognosis and significant efforts are being made to develop more effective treatment methods. Sonodynamic therapy (SDT) is an emerging modality for cancer treatment which combines ultrasound with sonosensitizers to produce a localized cytotoxic effect. It has long been known that ultrasound exposure can cause both thermal and non-thermal bioeffects and it remains an open question to what degree does temperature impact the efficacy of SDT. In order to optimize the ultrasound parameters of SDT, transcranial MRI-guided focused ultrasound (MRgFUS) and real-time MRI thermometry were used to monitor the therapy in a rat brain tumor model. Experiments were performed using a C6 intracranial glioma tumor model in 37 male Sprague Dawley rats. Treatments were performed about 7 days following tumor implantation when the tumor reached 1-3 mm in diameter as determined by MRI. 5-aminolevulinic acid (5-ALA) was injected at a dose of 60 mg/kg six hours before sonication. MRgFUS at 1.06 MHz was delivered continuously at an in situ spatial-peak temporal-average intensity of 5.5 W/cm2 for 20 min. MR thermometry was acquired to monitor the temperature change in the brain during sonication. The tumor growth response for animals receiving 5-ALA alone, FUS alone, 5-ALA + FUS and a sham control group were evaluated with MRI every week following treatment. During 20 min of MRgFUS at 5.5 W/cm2, the temperature within the targeted brain tumor was elevated from 32.3 ± 0.5 °C and 37.2 ± 0.7 °C to 33.2 ± 0.9 °C and 38.4 ± 1.1 °C, respectively. Both the tumor growth inhibition and survival were significantly improved in the 5-ALA + FUS group with 32 °C or 37 °C as the starting core body (rectal) temperature. 5-ALA alone and FUS alone did not improve survival. These promising results indicate that relatively low power continuous wave transcranial MRgFUS in conjunction with 5-ALA can produce an inhibitory effect on rat brain tumor growth in the absence of thermal dose. Further investigation of the ultrasound parameters is needed to improve the therapeutic efficacy of MRgFUS and 5-ALA.


Asunto(s)
Neoplasias Encefálicas/terapia , Modelos Animales de Enfermedad , Glioma/terapia , Ácidos Levulínicos/farmacología , Radioterapia Guiada por Imagen/métodos , Terapia por Ultrasonido/métodos , Animales , Neoplasias Encefálicas/patología , Glioma/patología , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas , Ácido Aminolevulínico
13.
Sci Rep ; 8(1): 9013, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899537

RESUMEN

Leptomeningeal metastases (LM) are a serious complication of cancer in the central nervous system (CNS) and are diagnosed in approximately 5% of patients with solid tumors. Effective treatment using systemically administered therapeutics is hindered by the barriers of the CNS. Ultrasound can mediate delivery of drugs through these barriers. The goal of this study was to test the feasibility of using ultrasound-mediated drug delivery to improve the treatment of LM. LM was induced in the spinal cord of athymic rats by injecting HER2-expressing breast cancer cells into the subarachnoid space of the thoracic spine. Animals were divided into three groups: no treatment (n = 5), trastuzumab only (n = 6) or trastuzumab + focused ultrasound + microbubbles (FUS + MBs) (n = 7). Animals in groups 2 and 3 were treated weekly with intravenous trastuzumab +/- FUS + MBs for three weeks. Suppression in tumor growth was qualitatively observed by MRI in the group receiving ultrasound, and was confirmed by a significant difference in the tumor volume measured from the histology data (25 ± 17 mm3 vs 8 ± 5 mm3, p = 0.04 in the trastuzumab-only vs trastuzumab + FUS + MBs). This pilot study demonstrates the potential of ultrasound-mediated drug delivery as a novel treatment for LM. Future studies will extend this work to larger cohorts and the investigation of LM arising from other cancers.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Neoplasias Meníngeas/terapia , Microburbujas , Trastuzumab/farmacología , Ultrasonografía/métodos , Animales , Antineoplásicos Inmunológicos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Imagen por Resonancia Magnética , Neoplasias Meníngeas/diagnóstico por imagen , Neoplasias Meníngeas/secundario , Proyectos Piloto , Ratas Desnudas , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos
14.
IEEE Trans Biomed Eng ; 64(12): 2880-2889, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28328497

RESUMEN

OBJECTIVE: Delivery barriers of nanodrug in large tumors due to heterogeneous blood supply, elevated interstitial pressure, and long transport distances can degrade the efficacy of cancer treatment. In this study, we proposed a therapeutic strategy to improve the tumor growth inhibition by injecting pegylated liposomal doxorubicin (PLD), and then applying a short time of ultrasound hyperthermia (HT) on the entire solid tumor, and inflicting ultrasound thermal ablation (Ab) in the low-perfused tumor region. METHODS: BALB/c female mice with an average weight of 20 g were adopted and murine breast cancer cells 4T1 were subcutaneously implanted into the flank. A 1.0-MHz planar and a 0.47-MHz focused ultrasound transducers were used, respectively, for the HT and Ab treatment. RESULTS: For a PLD dose of 5 mg/kg, the PLD + HT(42 °C, 10 min) group caused a significant decrease in the tumor size as compared with the control and the PLD group, but there were no significant differences between the PLD + HT group and the PLD + Ab(56 °C, 49 s) + HT group. For a PLD dose of 3 mg/kg, the tumor sizes among the four groups were mutually significant. The level of reduction in tumor was PLD + Ab + HT > PLD + HT > PLD > control. CONCLUSION: The combination of anticancer nanodrug and ultrasound thermal treatment could remarkably suppress cancer tumor growth with a minimum compromise of side effects. SIGNIFICANCE: The strategy of using thermal Ab in locations that are not reached by nanodrug with mild HT shows a promising potential for the entire tumor treatment.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Hipertermia Inducida/métodos , Neoplasias Mamarias Experimentales/terapia , Nanomedicina Teranóstica/métodos , Animales , Línea Celular Tumoral , Femenino , Histocitoquímica , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C
15.
Nanoscale Res Lett ; 12(1): 62, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28110445

RESUMEN

The use of photodynamic therapy (PDT) in the treatment of brain cancer has produced exciting results in clinical trials over the past decade. PDT is based on the concept that a photosensitizer exposed to a specific light wavelength produces the predominant cytotoxic agent, to destroy tumor cells. However, delivering an efficient light source to the brain tumor site is still a challenge. The light source should be delivered by placing external optical fibers into the brain at the time of surgical debulking of the tumor. Consequently, there exists the need for a minimally invasive treatment for brain cancer PDT. In this study, we investigated an attractive non-invasive option on glioma cell line by using Tb3+-doped LaF3 scintillating nanoparticles (LaF3:Tb) in combination with photosensitizer, meso-tetra(4-carboxyphenyl)porphyrin (MTCP), followed by activation with soft X-ray (80 kVp). Scintillating LaF3:Tb nanoparticles, with sizes of approximately 25 nm, were fabricated. The particles have a good dispersibility in aqueous solution and possess high biocompatibility. However, significant cytotoxicity was observed in the glioma cells while the LaF3:Tb nanoparticles with MTCP were exposed under X-ray irradiation. The study has demonstrated a proof of concept as a non-invasive way to treat brain cancer in the future.

16.
Ultrason Sonochem ; 36: 198-205, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28069201

RESUMEN

The clinical application of chemotherapeutics for brain tumors remains a challenge due to limitation of blood-brain barrier/blood-tumor barrier (BBB/BTB). In this study, we investigated the effects of low-dose focused ultrasound hyperthermia (UH) on the delivery and therapeutic efficacy of pegylated liposomal doxorubicin (PLD) for brain metastasis of breast cancer. Murine breast cancer cells (4T1-luc2) expressing firefly luciferase were implanted into mouse striatum as a brain tumor model. The mice were intravenously injected with PLD with/without transcranial pulsed-wave/continuous-wave UH (pUH/cUH) treatment on day-6 after tumor implantation. pUH (frequency: 500kHz, PRF: 1000Hz, duty cycle: 50%) was conducted under equal acoustic power (2.2-Watt) and sonication duration (10-min) as cUH. The amounts of doxorubicin accumulated in the normal brain and tumor tissues were measured with fluorometry. The tumor growth responses for the control, pUH, PLD, PLD+cUH, and PLD+pUH groups were evaluated with IVIS. The PLD distribution and cell apoptosis were assessed with immunofluorescence staining. The results showed that pUH significantly enhanced the PLD delivery into brain tumors and the tumor growth was further inhibited by PLD+pUH without damaging the sonicated normal brain tissues. This indicates that low-dose transcranial pUH is a promising method to selectively enhance nanodrug delivery and improve the brain tumor treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Sistemas de Liberación de Medicamentos/métodos , Hipertermia Inducida , Ondas Ultrasónicas , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Ratones , Nanoestructuras/química , Polietilenglicoles/química , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico
17.
Sci Rep ; 7: 46689, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28425493

RESUMEN

Microbubbles (MBs) serve as a critical catalyst to amplify local cavitation in CNS capillary lumen to facilitate focused ultrasound (FUS) to transiently open the blood-brain barrier (BBB). However, limited understanding is available regarding the effect of different microbubbles to induce BBB opening. The aim of this study is to characterize different MBs on their effect in FUS-induced BBB opening. Three MBs, SonoVue, Definity, and USphere, were tested, with 0.4-MHz FUS exposure at 0.62-1.38 of mechanical index (MI) on rats. Evans blue, dynamic contrast-enhanced (DCE) MRI and small-animal ultrasound imaging were used as surrogates to allow molecule-penetrated quantification, BBB-opened observation, and MBs circulation/persistence. Cavitation activity was measured via the passive cavitation detection (PCD) setup to correlate with the exposure level and the histological effect. Under given and identical MB concentrations, the three MBs induced similar and equivalent BBB-opening effects and persistence. In addition, a treatment paradigm by adapting exposure time is proposed to compensate MB decay to retain the persistence of BBB-opening efficiency in multiple FUS exposures. The results potentially improve understanding of the equivalence among MBs in focused ultrasound CNS drug delivery, and provide an effective strategy for securing persistence in this treatment modality.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Medios de Contraste/administración & dosificación , Microburbujas , Terapia por Ultrasonido/métodos , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Masculino , Ratas Sprague-Dawley
19.
J Control Release ; 211: 53-62, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26047759

RESUMEN

The clinical application of chemotherapy for brain cancer tumors remains a challenge due to difficulties in the transport of therapeutic agents across the blood-brain barrier/blood-tumor barrier (BBB/BTB). In this study, we developed des-octanoyl ghrelin-conjugated microbubbles (GMB) loaded with TGFß1 inhibitor (LY364947) (GMBL) to induce BBB/BTB disruption for ultrasound (US) sonication with GMBL. The in-vitro stability study showed that GMB was pretty stable over one month. The in-vivo study showed that the accumulation of superparamagnetic iron oxide nanoparticles (SPION) in the sonicated tumor was significantly higher for focused US sonication in the presence of GMBL, indicating that GMBL/US can locally disrupt BBB/BTB to promote vascular permeability of nanoparticles. In addition, the combination of folate-conjugated polymersomal doxorubicin (FPD) and GMBL/US (FPD+GMBL/US) achieved the best anti-glioma effect and significant improvement in the overall survival time for brain tumor-bearing mice. When combined with focused US, GMBL facilitated local BBB/BTB disruption and simultaneously released LY364947 to decrease the pericyte coverage of the endothelium at the targeted brain tumor sites, resulting in enhanced accumulation and antitumor activity of FPD. The overall results indicate that GMBL/US owns a great potential for non-invasive targeting delivery of nanomedicine across the BBB to treat central nervous system (CNS) diseases.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/terapia , Microburbujas , Nanomedicina/métodos , Sonicación/métodos , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Sistemas de Liberación de Medicamentos/métodos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones SCID , Pirazoles/administración & dosificación , Pirroles/administración & dosificación , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo
20.
PLoS One ; 9(2): e90107, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587228

RESUMEN

Erythropoietin (EPO) is a neuroprotective agent against cerebral ischemia/reperfusion (I/R)-induced brain injury. However, its crossing of blood-brain barrier is limited. Focused ultrasound (FUS) sonication with microbubbles (MBs) can effectively open blood-brain barrier to boost the vascular permeability. In this study, we investigated the effects of MBs/FUS on extending the therapeutic time window of EPO and its neuroprotective effects in both acute and chronic phases. Male Wistar rats were firstly subjected to two common carotid arteries and right middle cerebral artery occlusion (three vessels occlusion, 3VO) for 50 min, and then the rats were treated with hEPO (human recombinant EPO, 5000 IU/kg) with or without MBs/FUS at 5 h after occlusion/reperfusion. Acute phase investigation (I/R, I/R+MBs/FUS, I/R+hEPO, and I/R+hEPO+MBs/FUS) was performed 24 h after I/R; chronic tests including cylinder test and gait analysis were performed one month after I/R. The experimental results showed that MBs/FUS significantly increased the cerebral content of EPO by bettering vascular permeability. In acute phase, both significant improvement of neurological score and reduction of infarct volume were found in the I/R+hEPO+MBs/FUS group, as compared with I/R and I/R+hEPO groups. In chronic phase, long-term behavioral recovery and neuronal loss in brain cortex after I/R injury was significantly improved in the I/R+hEPO+MBs/FUS group. This study indicates that hEPO administration with MBs/FUS sonication even at 5 h after occlusion/reperfusion can produce a significant neuroprotection.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Eritropoyetina/administración & dosificación , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Daño por Reperfusión/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/patología , Sistemas de Liberación de Medicamentos/instrumentación , Marcha/efectos de los fármacos , Infarto de la Arteria Cerebral Media , Masculino , Microburbujas , Neuronas/patología , Ratas , Ratas Wistar , Daño por Reperfusión/patología , Sonido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA