Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nanoscale ; 12(4): 2773-2786, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31957767

ABSTRACT

Until now, magnetic hyperthermia was used to remove solid tumors by targeting magnetic nanoparticles (MNPs) to tumor sites. In this study, leukemia cells in the bloodstream were directly removed by whole-body hyperthermia, using leukemia cell-specific MNPs. An epithelial cellular adhesion molecule (EpCAM) antibody was immobilized on the surface of MNPs (EpCAM-MNPs) to introduce the specificity of MNPs to leukemia cells. The viability of THP1 cells (human monocytic leukemia cells) was decreased to 40.8% of that in control samples by hyperthermia using EpCAM-MNPs. In AKR mice, an animal model of lymphoblastic leukemia, the number of leukemia cells was measured following the intravenous injection of EpCAM-MNPs and subsequent whole-body hyperthermia treatment. The result showed that the leukemia cell number was also decreased to 43.8% of that without the treatment of hyperthermia, determined by Leishman staining of leukemia cells. To support the results, simulation analysis of heat transfer from MNPs to leukemia cells was performed using COMSOL Multiphysics simulation software. The surface temperature of leukemia cells adhered to EpCAM-MNPs was predicted to be increased to 82 °C, whereas the temperature of free cells without adhered MNPs was predicted to be 38 °C. Taken together, leukemia cells were selectively removed by magnetic hyperthermia from the bloodstream, because EpCAM-modified magnetic particles were specifically attached to leukemia cell surfaces. This approach has the potential to remove metastatic cancer cells, and pathogenic bacteria and viruses floating in the bloodstream.


Subject(s)
Hyperthermia, Induced/methods , Magnetite Nanoparticles/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antibodies, Immobilized/administration & dosage , Antibodies, Immobilized/chemistry , Cell Line , Cell Survival , Disease Models, Animal , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cell Adhesion Molecule/metabolism , Humans , Immunomagnetic Separation , Magnetite Nanoparticles/chemistry , Mice , Mice, Inbred AKR , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
2.
J Biomater Sci Polym Ed ; 26(16): 1178-89, 2015.
Article in English | MEDLINE | ID: mdl-26249213

ABSTRACT

A tumor-targeted and pH-responsive drug release system based on superparamagnetic iron oxide nanoparticles (IONPs) coated by poly(ethylene glycol) (PEG) and dodecylamine (DDA)-modified polyitaconic acid (PIA) connecting with bortezomib (BTZ) (PIA-PEG-DDA-BTZ@IOs) has been constructed and characterized. The anticancer drug BTZ was first conjugated using dopamine as the linker via catechol borate ester bond, which is acid cleavable and used as an ideal pH-responsive drug release system. The IONPs were then coated by PIA-PEG-DDA-BTZ to form micelles with good biocompatibility. The conjugates were further designed to target liver cancer cells overexpressing vascular endothelial growth factor (VEGF) by the targeting molecule anti-vascular endothelial growth factor (anti-VEGF). The magnetic resonance imaging showed that the targeting capability of IONPs-anti-VEGF conjugates to Hep G2 cells was more significant than that of non-anti-VEGF IONPs. From the above, this kind of novel dual-functional targeting probe could provide a new idea for the diagnosis and treatment of cancer.


Subject(s)
Antibodies, Immobilized/administration & dosage , Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Liver Neoplasms, Experimental/metabolism , Magnetite Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Absorption, Physiological , Amines/chemistry , Animals , Antibodies, Immobilized/chemistry , Antibodies, Immobilized/metabolism , Antibodies, Immobilized/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Bortezomib/administration & dosage , Bortezomib/metabolism , Bortezomib/pharmacokinetics , Bortezomib/pharmacology , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/metabolism , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Liberation , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hydrogen-Ion Concentration , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Magnetite Nanoparticles/ultrastructure , Male , Mice, Inbred ICR , Polyethylene Glycols/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Succinates/chemistry , Surface Properties , Tissue Distribution , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
3.
J Biomater Appl ; 30(5): 558-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26184354

ABSTRACT

Recently, we have shown that anti-BMP2 monoclonal antibodies (mAbs) can trap endogenous osteogenic BMP ligands, which can in turn mediate osteodifferentiation of progenitor cells. The effectiveness of this strategy requires the availability of the anti-BMP-2 monoclonal antibodies antigen-binding sites for anti-BMP-2 monoclonal antibodies to bind to the scaffold through a domain that will leave its antigen-binding region exposed and available for binding to an osteogenic ligand. We examined whether antibodies bound to a scaffold by passive adsorption versus through Protein G as a linker will exhibit differences in mediating bone formation. In vitro anti-BMP-2 monoclonal antibodies was immobilized on absorbable collagen sponge (ACS) with Protein G as a linker to bind the antibody through its Fc region and implanted into rat calvarial defects. The biomechanical strength of bone regenerated by absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies immune complex was compared to ACS/anti-BMP-2 monoclonal antibodies or ACS/Protein G/isotype mAb control group. Results demonstrated higher binding of anti-BMP-2 monoclonal antibodies/BMPs to C2C12 cells, when the mAb was initially attached to recombinant Protein G or Protein G-coupled microbeads. After eight weeks, micro-CT and histomorphometric analyses revealed increased bone formation within defects implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies compared with defects implanted with absorbable collagen sponge/anti-BMP-2 monoclonal antibodies (p < 0.05). Confocal laser scanning microscopy (CLSM) confirmed increased BMP-2, -4, and -7 detection in sites implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies in vivo. Biomechanical analysis revealed the regenerated bone in sites with Protein G/anti-BMP-2 monoclonal antibodies had higher mechanical strength in comparison to anti-BMP-2 monoclonal antibodies. The negative control group, Protein G/isotype mAb, did not promote bone regeneration and exhibited significantly lower mechanical properties (p < 0.05). Altogether, our results demonstrated that application of Protein G as a linker to adsorb anti-BMP-2 monoclonal antibodies onto the scaffold was accompanied by increased in vitro binding of the anti-BMP-2 mAb/BMP immune complex to BMP-receptor positive cell, as well as increased volume and strength of de novo bone formation in vivo.


Subject(s)
Absorbable Implants , Antibodies, Immobilized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Bone Morphogenetic Protein 2/immunology , Bone Regeneration/drug effects , Skull/drug effects , Skull/physiology , Animals , Antibodies, Immobilized/administration & dosage , Antibodies, Immobilized/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Cell Line , Collagen/chemistry , Female , Mice , Rats, Sprague-Dawley , Skull/immunology , Skull/injuries
4.
J Cardiovasc Pharmacol Ther ; 20(3): 322-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25412893

ABSTRACT

Antibody-coated stents to capture circulating endothelial progenitor cells (EPCs) for re-endothelialization appear to be a novel therapeutic option for the treatment of atherosclerotic disease. Hydroxybutyl chitosan (HBC), a linear polysaccharide made from shrimps and other crustacean shells, is biocompatible, nontoxic, and hydrophilic, making it ideal for biomedical applications. In this study, HBC was explored for the immobilization of anti-CD133 antibodies. We demonstrated that CD133 antibodies mediated by HBC were successfully coated on cobalt-chromium alloy discs and metal stents. The coating was homogeneous and smooth as shown by electronic microscopy analysis. Balloon expansion of coated stents did not cause cracking or peeling. The HBC discs promoted CD133+ EPCs and human umbilical vein endothelial cell growth in vitro. The CD133 antibody-coated but not bare discs bound CD133+ EPCs in vitro. Implantation of CD133 antibody-coated stents significantly inhibited intimal hyperplasia and reduced restenosis compared with implantation of bare stents in a porcine model of atherosclerosis. These findings suggest HBC is a valuable anchoring agent that can be applied for bioactive coating of stents and that CD133 antibody-coated stents might be a potential therapeutic alternative for the treatment of atherosclerotic disease.


Subject(s)
Antibodies, Immobilized/administration & dosage , Antigens, CD/immunology , Atherosclerosis/therapy , Chitosan/administration & dosage , Endothelial Progenitor Cells/physiology , Glycoproteins/immunology , Peptides/immunology , Polymers/administration & dosage , Stents , AC133 Antigen , Animals , Cell Proliferation , Cells, Cultured , Humans , Hyperplasia , Male , Neointima/pathology , Swine
5.
Curr Cancer Drug Targets ; 13(1): 11-29, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23030233

ABSTRACT

Pulmonary delivery of drug-loaded nanoparticles is a novel approach for lung cancer treatment and the conjugation of nanoparticles to a targeting ligand further promotes specificity of the carrier cargo to cancer cells. Notably, the epithelial cell adhesion molecule (EpCAM, CD326) is over expressed in lung cancer. Here, we report the safety and proof-of-concept efficacy of drug-loaded nanoparticles and EpCAM immunonanoparticles in a c-Raf transgenic lung cancer model. PEG-PLA nanoparticles and immunonanoparticles were prepared whereby paclitaxel palmitate (Pcpl) was incorporated as a medication for its common use in lung cancer treatment. Four doses of aerosolized nanoparticle formulations or vehicle were endotracheally administered to mice by consecutive or alternate regimes. Pulmonary delivery of drug loaded nano- and/or immunonanoparticle formulations elicited mild inflammation as evidenced by the slightly increased neutrophil and activated macrophage counts in bronchoalveolar lavage. No evidence for pulmonary toxicity following treatment with either blank or drug-loaded nano- and/or immunonanoparticles was observed. Proof-of-concept efficacy was determined by serial CT scanning and histopathology. Animals treated with either EpCAM antibody or Pcpl solution or drug loaded nano- or immunonanoparticles inhibited disease progression. Conversely, disease progression was noted with vehicle treated animals with nearly 30% loss of their aerated lung volume. Importantly, treatment of mice with either Pcpl or EpCAM antibody solution caused 80% mortality and/or haemorrhage, respectively, thus causing unacceptable toxicity. In contrast, the survival of animals treated with either nano- or immunonanoparticles was 60 and 70%, respectively. Taken collectively, pulmonary delivered drug-loaded nano- and EpCAM immunonanoparticles were well tolerated and can be considered a promising strategy for improving lung cancer treatment.


Subject(s)
Antibodies, Immobilized/administration & dosage , Antibodies, Neoplasm/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Adhesion Molecules/antagonists & inhibitors , Disease Models, Animal , Drug Delivery Systems , Lung Neoplasms/drug therapy , Administration, Inhalation , Animals , Antibodies, Immobilized/adverse effects , Antibodies, Immobilized/therapeutic use , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/adverse effects , Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Adhesion Molecules/metabolism , Drug Delivery Systems/adverse effects , Epithelial Cell Adhesion Molecule , Female , Humans , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Nanoparticles/administration & dosage , Nanoparticles/adverse effects , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/analogs & derivatives , Paclitaxel/therapeutic use , Palmitates/administration & dosage , Palmitates/adverse effects , Palmitates/therapeutic use , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Random Allocation , Surface Properties , Survival Analysis
6.
Nanomedicine (Lond) ; 7(5): 691-703, 2012 May.
Article in English | MEDLINE | ID: mdl-22630152

ABSTRACT

BACKGROUND: The prostate stem cell antigen (PSCA) is broadly overexpressed on the surface of prostate cancer cells. MATERIALS & METHODS: Anti-human PSCA monoclonal antibody (mAb 7F5) was bound to Fe(3)O(4)/Au (GoldMag) nanoparticles to serve as a PSCA-specific molecular MRI probe (mAb 7F5@GoldMag) for in vivo detection of prostate cancer cells. First, the efficacy of the antibody immobilization for the binding was assessed. Next, PC-3 (human prostate cancer cell line with PSCA overexpression) tumor-bearing mice were injected with mAb 7F5@GoldMag for MRI measurements while using mouse anti-human IgG bound to the particles (IgG@GoldMag) to serve as a nonspecific control. MRI examinations were conducted before and after injection of these probes at 6, 12 and 24 h; T2-weighted signal intensity within the tumors was measured. RESULTS: Targeted binding of the mAb 7F5@GoldMag probe to PC-3 tumors was verified with optical images and MRI; selective binding was not observed for the nonspecific IgG@GoldMag probe. CONCLUSION: MRI measurements suggest the promising efficacy of this new approach for targeted molecular imaging of prostate tumors.


Subject(s)
Antigens, Neoplasm , Magnetic Resonance Imaging/methods , Metal Nanoparticles , Neoplasm Proteins , Prostatic Neoplasms , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/chemistry , Antibodies, Immobilized/administration & dosage , Antibodies, Immobilized/chemistry , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/immunology , Gold/administration & dosage , Gold/chemistry , Humans , Male , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Mice , Molecular Imaging , Molecular Targeted Therapy , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Neoplastic Stem Cells/diagnostic imaging , Neoplastic Stem Cells/immunology , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/immunology , Radiography
7.
J Am Chem Soc ; 132(20): 6906-7, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20433206

ABSTRACT

We report that antibodies can be spontaneously loaded in functionalized mesoporous silica (FMS) with superhigh density (0.4-0.8 mg of antibody/mg of FMS) due to their comprehensive noncovalent interaction. The superhigh loading density and noncovalent interaction between FMS and antibodies allow long-lasting local release of the immunoregulatory molecules from FMS under physiological conditions. Preliminary data indicate that FMS-anti-CTLA4 antibody injected directly into a mouse melanoma induces much greater and extended inhibition of tumor growth than the antibody given systemically. Our findings open up a novel approach for local delivery of therapeutically active proteins to tumors and, potentially, other diseases.


Subject(s)
Antibodies, Immobilized/administration & dosage , Antibodies, Immobilized/therapeutic use , Immunotherapy , Nanostructures/chemistry , Neoplasms/therapy , Animals , Antibodies, Immobilized/chemistry , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/therapeutic use , Mice , Porosity , Rats , Silicon Dioxide/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...