Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 1655, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733537

ABSTRACT

Improvements in dimensional metrology and innovations in physical-chemical characterization of functionalized nanoparticles are critically important for the realization of enhanced performance and benefits of nanomaterials. Toward this goal, we propose a multi-technique measurement approach, in which correlated atomic force microscopy, dynamic light scattering, high performance liquid chromatography and mass spectroscopy measurements are used to assess molecular and structural properties of self-assembled polyplex nanoparticles with a core-shell structure. In this approach, measurement methods are first validated with a model system consisting of gold nanoparticles functionalized with synthetic polycationic branched polyethylenimine macromolecules. Shell thickness is measured by atomic force microscopy and dynamic light scattering, and the polyelectrolyte uptake determined by chromatographic separation and mass spectrometric analysis. Statistical correlation between size, structure and stability provide a basis for extending the methods to more complex self-assembly of nucleic acids and macromolecules via a condensation reaction. From these size and analytical chemical measurements, we obtain a comprehensive spatial description of these assemblies, obtain a detailed interpretation of the core-shell evolution, and identify regions of the parameter space where stable, discrete particle formation occurs.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Microscopy, Atomic Force/methods , Polyelectrolytes/chemistry , Polyethyleneimine/chemistry , Polymers/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Surface Properties
2.
ACS Nano ; 8(6): 5494-514, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24811110

ABSTRACT

Temozolomide (TMZ)-resistance in glioblastoma multiforme (GBM) has been linked to upregulation of O(6)-methylguanine-DNA methyltransferase (MGMT). Wild-type (wt) p53 was previously shown to down-modulate MGMT. However, p53 therapy for GBM is limited by lack of efficient delivery across the blood brain barrier (BBB). We have developed a systemic nanodelivery platform (scL) for tumor-specific targeting (primary and metastatic), which is currently in multiple clinical trials. This self-assembling nanocomplex is formed by simple mixing of the components in a defined order and a specific ratio. Here, we demonstrate that scL crosses the BBB and efficiently targets GBM, as well as cancer stem cells (CSCs), which have been implicated in recurrence and treatment resistance in many human cancers. Moreover, systemic delivery of scL-p53 down-modulates MGMT and induces apoptosis in intracranial GBM xenografts. The combination of scL-p53 and TMZ increased the antitumor efficacy of TMZ with enhanced survival benefit in a mouse model of highly TMZ-resistant GBM. scL-p53 also sensitized both CSCs and bulk tumor cells to TMZ, increasing apoptosis. These results suggest that combining scL-p53 with standard TMZ treatment could be a more effective therapy for GBM.


Subject(s)
Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Nanoparticles/chemistry , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Blood-Brain Barrier/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Genetic Therapy/methods , Glioblastoma/genetics , Humans , Mice , Microscopy, Atomic Force , Nanomedicine , Neoplasm Recurrence, Local , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Temozolomide , Treatment Outcome
3.
Nanomedicine ; 4(4): 318-29, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18676207

ABSTRACT

To circumvent the problem of reduction of the supermagnetic properties of superparamagnetic iron oxide (SPIO) nanoparticles after chemical modification to conjugate targeting molecules, we have adapted a tumor-targeting nanoimmunoliposome platform technology (scL) to encapsulate and deliver SPIO (scL-SPIO) in vitro and in vivo without chemical modification. Scanning probe microscopy, confocal microscopy, and Prussian blue staining were used to analyze the scL-SPIO and assess intracellular uptake and distribution of SPIO in vitro. In vivo targeting and tumor-specific uptake of scL-SPIO was examined using fluorescent-labeled SPIO. We demonstrated that SPIO encapsulation in the scL complex results in an approximately 11-fold increase in SPIO uptake in human cancer cells in vitro, with distribution to cytoplasm and nucleus. Moreover, the scL nanocomplex specifically and efficiently delivered SPIO into tumor cells after systemic administration, demonstrating the potential of this approach to enhance local tumor concentration and the utility of SPIO for clinical applications.


Subject(s)
Breast Neoplasms/pathology , Drug Delivery Systems/methods , Ferric Compounds/administration & dosage , Nanoparticles/chemistry , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Female , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Humans , Image Enhancement/methods , Liposomes/chemistry , Magnetic Resonance Imaging/methods , Microscopy, Confocal , Microscopy, Scanning Probe , Molecular Structure , Spectroscopy, Fourier Transform Infrared
4.
Cancer Res ; 67(7): 2938-43, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409398

ABSTRACT

The field of small interfering RNA (siRNA) as potent sequence-selective inhibitors of transcription is rapidly developing. However, until now, low transfection efficiency, poor tissue penetration, and nonspecific immune stimulation by in vivo administered siRNAs have delayed their therapeutic application. Their potential as anticancer therapeutics hinges on the availability of a vehicle that can be systemically administered, safely and repeatedly, and will deliver the siRNA specifically and efficiently to the tumor, both primary tumors and metastases. We have developed a nanosized immunoliposome-based delivery complex (scL) that, when systemically administered, will preferentially target and deliver molecules useful in gene medicine, including plasmid DNA and antisense oligonucleotides, to tumor cells wherever they occur in the body. This tumor-targeting nanoparticle delivery vehicle can also deliver siRNA to both primary and metastatic disease. We have also enhanced the efficiency of this complex by the inclusion of a pH-sensitive histidine-lysine peptide in the complex (scL-HoKC) and by delivery of a modified hybrid (DNA-RNA) anti-HER-2 siRNA molecule. Scanning probe microscopy confirms that this modified complex maintains its nanoscale size. More importantly, we show that this nanoimmunoliposome anti-HER-2 siRNA complex can sensitize human tumor cells to chemotherapeutics, silence the target gene and affect its downstream pathway components in vivo, and significantly inhibit tumor growth in a pancreatic cancer model. Thus, this complex has the potential to help translate the potent effects of siRNA into a clinically viable anticancer therapeutic.


Subject(s)
Breast Neoplasms/therapy , Genetic Therapy/methods , Liposomes/administration & dosage , Pancreatic Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Mice , Mice, Nude , Nanoparticles/administration & dosage , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...