Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 212: 1-9, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26071631

ABSTRACT

Focused ultrasound (FUS) exposure in the presence of microbubbles can temporally open the blood-brain barrier (BBB) and is an emerging technique for non-invasive brain therapeutic agent delivery. Given the potential to deliver large molecules into the CNS via this technique, we propose a reliable strategy to synergistically apply FUS-BBB opening for the non-invasive and targeted delivery of non-viral genes into the CNS for therapeutic purpose. In this study, we developed a gene-liposome system, in which the liposomes are designed to carry plasmid DNA (pDNA, containing luciferase reporter gene) to form a liposomal-plasmid DNA (LpDNA) complex. Pulsed FUS exposure was delivered to induce BBB opening (500-kHz, burst length=10ms, 1% duty cycle, PRF=1Hz). The longitudinal expression of luciferase was quantitated via an in vivo imaging system (IVIS). The reporter gene expression level was confirmed via immunoblotting, and histological staining was used to identify transfected cells via fluorescent microscopy. In a comparison of gene transduction efficiency, the LpDNA system showed better cell transduction than the pDNA system. With longitudinal observation of IVIS monitoring, animals with FUS treatment showed significant promotion of LpDNA release into the CNS and demonstrated enhanced expression of genes upon sonication with FUS-BBB opening, while both the luciferase and GDNF protein expression were successfully measured via Western blotting. The gene expression peak was observed at day 2, and the gene expression level was up to 5-fold higher than that in the untreated hemisphere (compared to a 1-fold increase in the direct-inject positive-control group). The transfection efficiency was also found to be LpDNA dose-dependent, where higher payloads of pDNA resulted in a higher transfection rate. Immunoblotting and histological staining confirmed the expression of reporter genes in glial cells as well as astrocytes. This study suggests that IV administration of LpDNA in combination with FUS-BBB opening can provide effective gene delivery and expression in the CNS, demonstrating the potential to achieve non-invasive and targeted gene delivery for treatment of CNS diseases.


Subject(s)
Blood-Brain Barrier/metabolism , DNA/administration & dosage , Gene Transfer Techniques , Microbubbles , Ultrasonic Waves , Animals , Astrocytes/metabolism , Genes, Reporter , Glial Cell Line-Derived Neurotrophic Factor/genetics , Liposomes , Luciferases/genetics , Male , Mice, Inbred BALB C , Neuroglia/metabolism , Plasmids
2.
PLoS One ; 8(3): e58995, 2013.
Article in English | MEDLINE | ID: mdl-23527068

ABSTRACT

The purpose of this study is to assess the preclinical therapeutic efficacy of magnetic resonance imaging (MRI)-monitored focused ultrasound (FUS)-induced blood-brain barrier (BBB) disruption to enhance Temozolomide (TMZ) delivery for improving Glioblastoma Multiforme (GBM) treatment. MRI-monitored FUS with microbubbles was used to transcranially disrupt the BBB in brains of Fisher rats implanted with 9L glioma cells. FUS-BBB opening was spectrophotometrically determined by leakage of dyes into the brain, and TMZ was quantitated in cerebrospinal fluid (CSF) and plasma by LC-MS\MS. The effects of treatment on tumor progression (by MRI), animal survival and brain tissue histology were investigated. Results demonstrated that FUS-BBB opening increased the local accumulation of dyes in brain parenchyma by 3.8-/2.1-fold in normal/tumor tissues. Compared to TMZ alone, combined FUS treatment increased the TMZ CSF/plasma ratio from 22.7% to 38.6%, reduced the 7-day tumor progression ratio from 24.03 to 5.06, and extended the median survival from 20 to 23 days. In conclusion, this study provided preclinical evidence that FUS BBB-opening increased the local concentration of TMZ to improve the control of tumor progression and animal survival, suggesting its clinical potential for improving current brain tumor treatment.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/radiation effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Sound , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Brain/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Cell Line, Tumor , Dacarbazine/pharmacokinetics , Dacarbazine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Glioblastoma/diagnosis , Glioblastoma/mortality , Magnetic Resonance Imaging , Male , Rats , Temozolomide
3.
J Magn Reson Imaging ; 34(6): 1313-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21965168

ABSTRACT

PURPOSE: To verify that low-frequency planar ultrasound can be used to disrupt the BBB in large animals, and the usefulness of MRI to quantitatively monitor the delivery of superparamagnetic iron oxide (SPIO) nanoparticles into the disrupted regions. MATERIALS AND METHODS: Two groups of swine subjected to craniotomy were sonicated with burst lengths of 30 or 100 ms, and one group of experiment was also performed to confirm the ability of 28-kHz sonication to open the BBB transcranially. SPIO nanoparticles were administered to the animals after BBB disruption. Procedures were monitored by MRI; SPIO concentrations were estimated by relaxivity mapping. RESULTS: Sonication for 30 ms created shallow disruptions near the probe tip; 100-ms sonications after craniotomy can create larger and more penetrating openings, increasing SPIO leakage ∼3.6-fold than 30-ms sonications. However, this was accompanied by off-target effects possibly caused by ultrasonic wave reflection. SPIO concentrations estimated from transverse relaxation rate maps correlated well with direct measurements of SPIO concentration by optical emission spectrometry. We have also shown that transcranial low-frequency 28-kHz sonication can induce secure BBB opening from longitudinal MR image follow up to 7 days. CONCLUSION: This study provides valuable information regarding the use low-frequency ultrasound for BBB disruption and suggest that SPIO nanoparticles has the potential to serve as a thernostic agent in MRI-guided ultrasound-enhanced brain drug delivery.


Subject(s)
Blood-Brain Barrier/physiology , Contrast Media/analysis , Magnetic Resonance Imaging , Magnetite Nanoparticles/analysis , Sonication/methods , Animals , Brain Chemistry , Craniotomy , Male , Nanoparticles , Swine
4.
Biomaterials ; 32(2): 516-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21030073

ABSTRACT

This study describes the creation and characterization of drug carriers prepared using the polymer poly[aniline-co-N-(1-one-butyric acid) aniline] (SPAnH) coated on Fe(3)O(4) cores to form three types of magnetic nanoparticles (MNPs); these particles were used to enhance the therapeutic capacity and improve the thermal stability of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), a compound used to treat brain tumors. The average hydrodynamic diameter of the MNPs was 89.2 ± 8.5 nm and all the MNPs displayed superparamagnetic properties. A maximum effective dose of 379.34 µg BCNU could be immobilized on 1 mg of MNP-3 (bound-BCNU-3). Bound-BCNU-3 was more stable than free-BCNU when stored at 4 °C, 25 °C or 37 °C. Bound-BCNU-3 could be concentrated at targeted sites in vitro and in vivo using an externally applied magnet. When applied to brain tumors, magnetic targeting increased the concentration and retention of bound-BCNU-3. This drug delivery system promises to provide more effective tumor treatment using lower therapeutic doses and potentially reducing the side effects of chemotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carmustine/therapeutic use , Drug Delivery Systems/methods , Glioma/drug therapy , Nanoparticles/chemistry , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Carmustine/administration & dosage , Carmustine/chemistry , Cell Line , Cell Line, Tumor , Glioma/pathology , Glioma/ultrastructure , Humans , Magnetic Resonance Imaging , Male , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley
5.
Proc Natl Acad Sci U S A ; 107(34): 15205-10, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696897

ABSTRACT

The superparamagnetic properties of magnetic nanoparticles (MNPs) allow them to be guided by an externally positioned magnet and also provide contrast for MRI. However, their therapeutic use in treating CNS pathologies in vivo is limited by insufficient local accumulation and retention resulting from their inability to traverse biological barriers. The combined use of focused ultrasound and magnetic targeting synergistically delivers therapeutic MNPs across the blood-brain barrier to enter the brain both passively and actively. Therapeutic MNPs were characterized and evaluated both in vitro and in vivo, and MRI was used to monitor and quantify their distribution in vivo. The technique could be used in normal brains or in those with tumors, and significantly increased the deposition of therapeutic MNPs in brains with intact or compromised blood-brain barriers. Synergistic targeting and image monitoring are powerful techniques for the delivery of macromolecular chemotherapeutic agents into the CNS under the guidance of MRI.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Blood-Brain Barrier , Brain Neoplasms/blood supply , Brain Neoplasms/ultrastructure , Contrast Media , Epirubicin/administration & dosage , Epirubicin/therapeutic use , Magnetic Resonance Imaging , Magnetics , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley , Ultrasonic Therapy
6.
Neuro Oncol ; 12(10): 1050-60, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20663792

ABSTRACT

Malignant glioma is a common and severe primary brain tumor with a high recurrence rate and an extremely high mortality rate within 2 years of diagnosis, even when surgical, radiological, and chemotherapeutic interventions are applied. Intravenously administered drugs have limited use because of their adverse systemic effects and poor blood-brain barrier penetration. Here, we combine 2 methods to increase drug delivery to brain tumors. Focused ultrasound transiently permeabilizes the blood-brain barrier, increasing passive diffusion. Subsequent application of an external magnetic field then actively enhances localization of a chemotherapeutic agent immobilized on a novel magnetic nanoparticle. Combining these techniques significantly improved the delivery of 1,3-bis(2-chloroethyl)-1-nitrosourea to rodent gliomas. Furthermore, the physicochemical properties of the nanoparticles allowed their delivery to be monitored by magnetic resonance imaging (MRI). The resulting suppression of tumor progression without damaging the normal regions of the brain was verified by MRI and histological examination. This noninvasive, reversible technique promises to provide a more effective and tolerable means of tumor treatment, with lower therapeutic doses and concurrent clinical monitoring.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Glioma/drug therapy , High-Energy Shock Waves/therapeutic use , Magnetics , Nanoparticles/administration & dosage , Animals , Blood-Brain Barrier/physiology , Carmustine/administration & dosage , Magnetic Resonance Imaging , Magnetics/methods , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...