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.
Nanomedicine (Lond) ; 11(5): 479-94, 2016.
Article in English | MEDLINE | ID: mdl-26892440

ABSTRACT

BACKGROUND: Delivery of PLGA (poly [D, L-lactide-co-glycolide])-based biodegradable nanoparticles (NPs) to antigen presenting cells, particularly dendritic cells, has potential for cancer immunotherapy. MATERIALS & METHODS: Using a PLGA NP vaccine construct CpG-NP-Tag (CpG-ODN-coated tumor antigen [Tag] encapsulating NP) prepared using solvent evaporation technique we tested the efficacy of ex vivo and in vivo use of this construct as a feasible platform for immune-based therapy. RESULTS: CpG-NP-Tag NPs were avidly endocytosed and localized in the endosomal compartment of bone marrow-derived dendritic cells. Bone marrow-derived dendritic cells exposed to CpG-NP-Tag NPs exhibited an increased maturation (higher CD80/86 expression) and activation status (enhanced IL-12 secretion levels). In vivo results demonstrated attenuation of tumor growth and angiogenesis as well as induction of potent cytotoxic T-lymphocyte responses. CONCLUSION: Collectively, results validate dendritic cells stimulatory response to CpG-NP-Tag NPs (ex vivo) and CpG-NP-Tag NPs' tumor inhibitory potential (in vivo) for therapeutic applications, respectively.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/drug effects , Nanoparticles/administration & dosage , Neoplasms/therapy , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Dendritic Cells/immunology , Endocytosis/immunology , Humans , Immunotherapy/methods , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Molecular Targeted Therapy , Nanoparticles/chemistry , Neoplasms/immunology , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
2.
Nanomedicine (Lond) ; 10(6): 915-29, 2015.
Article in English | MEDLINE | ID: mdl-25867857

ABSTRACT

AIM: Low immunogenicity remains a major obstacle in realizing the full potential of cancer vaccines. In this study, we evaluated CpG-coated tumor antigen (Tag)-encapsulating 'bacteriomimetic' nanoparticles (CpG-nanoparticle [NP]-Tag NPs) as an approach to enhance anti-tumor immunity. MATERIALS & METHODS: CpG-NP-Tag NPs were synthesized, characterized for their physicochemical properties and tested in vivo. RESULTS: We found CpG predosing followed by intraperitoneal (IP) immunization with CpG-NP-Tag NPs significantly attenuated tumor growth in female BALB/c mice compared with respective controls. Histopathological and Immunofluorescence data revealed CpG-NP-Tag tumors had lower proliferation, higher apoptotic activity, greater CD4(+) and CD8(+) T cell infiltration as well as higher IFN-γ levels as compared with control groups. CONCLUSION: Our findings suggest CpG-NP-Tag NPs can enhance anti-tumor effect of nanoparticulate tumor vaccination system.


Subject(s)
Antigens, Neoplasm/chemistry , Antineoplastic Agents/therapeutic use , Cancer Vaccines/immunology , CpG Islands/genetics , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Proliferation , Drug Delivery Systems , Female , Interferon-gamma/metabolism , Ki-67 Antigen/metabolism , Luminescence , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanomedicine/methods , Neoplasms/immunology , Neoplasms/therapy , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Spectrometry, Fluorescence
3.
In Vivo ; 26(5): 765-75, 2012.
Article in English | MEDLINE | ID: mdl-22949589

ABSTRACT

The phytoestrogens kaempferol, genistein and genistin were characterized using in vitro assays utilizing iodoacetic acid (IAA)-induced oxidative stress and mitochondrial dysfunction. RGC-5 cells were treated with different concentrations of IAA, and phytoestrogens were administered along with IAA. IAA is cytotoxic to RGC-5 cells and induces the generation of reactive oxygen species (ROS) in vitro. Genistein rescued RGC-5 cells in the presence of IAA, however, it also increased caspase activation and did not inhibit the generation of ROS. Genistein increased phosphorylation of ribosomal s6 kinase (p90RSK), reduced phosphorylation of the ribosomal S6 protein, and had no effect on phosphorylation of protein kinase B (AKT). Kaempferol and genistin rescued RGC-5 cells from IAA-induced cell death, as well as reduced caspase activation and ROS generation. Kaempferol increased phosphorylation of AKT and MAP kinase (p44/42). Genistin reduced phosphorylation of p42 and p90RSK. Although these phytoestrogens are flavonoids and similar in structure, they exhibit different effects on cell signaling.


Subject(s)
Apoptosis/drug effects , Free Radical Scavengers/pharmacology , Genistein/pharmacology , Isoflavones/pharmacology , Kaempferols/pharmacology , Oxidative Stress , Phytoestrogens/pharmacology , Animals , Apigenin/pharmacology , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Drug Stability , Enzyme Activation/drug effects , Estradiol/pharmacology , Flavonoids/pharmacology , Flavonols , Inhibitory Concentration 50 , Iodoacetic Acid/pharmacology , Membrane Potential, Mitochondrial/drug effects , Oxidants/pharmacology , Rats , Reactive Oxygen Species/metabolism
4.
PLoS One ; 7(9): e44299, 2012.
Article in English | MEDLINE | ID: mdl-22957061

ABSTRACT

Alternative survival pathways are commonly seen to be upregulated upon inhibition of receptor tyrosine kinases (RTK), including Her-2. It is established that treatment with Herceptin leads to selective overexpression and activation of epidermal growth factor receptor (EGFR) and Src which further contributes to oncogenesis in Herceptin resistant and triple negative breast cancer (TNBC) patients. Here, we show a co-regulated upregulation in the expression of Annexin A2 (AnxA2), a known substrate of Src and one of the regulators of EGFR receptor endocytosis, in Herceptin resistant and Her-2 negative breast cancer. Immunohistochemical expression analysis revealed a reciprocal regulation between Her-2 and AnxA2 in breast cancer clinical samples as well as in cell lines as confirmed by protein and RNA analysis. The siRNA and Herceptin mediated downregulation/inhibition of Her-2 in Her-2 amplified cells induced AnxA2 expression and membrane translocation. In this study we report a possible involvement of AnxA2 in maintaining constitutively activated EGFR downstream signaling intermediates and hence in cell proliferation, migration and viability. This effect was consistent in Herceptin resistant JIMT-1 cells as well as in Her-2 negative breast cancer. The siRNA mediated AnxA2 downregulation leads to increased apoptosis, decreased cell viability and migration. Our studies further indicate the role of AnxA2 in EGFR-Src membrane bound signaling complex and ligand induced activation of downstream signaling pathways. Targeting this AnxA2 dependent positive regulation of EGFR signaling cascade may be of therapeutic value in Her-2 negative breast cancer.


Subject(s)
Annexin A2/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Receptor, ErbB-2/metabolism , Apoptosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Survival , Collagen/chemistry , Drug Combinations , Drug Resistance, Neoplasm , Female , Humans , Immunohistochemistry/methods , Laminin/chemistry , Ligands , Phenotype , Proteoglycans/chemistry , Signal Transduction , Trastuzumab , src-Family Kinases/metabolism
5.
Biomaterials ; 33(29): 7164-73, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22795543

ABSTRACT

Delivery of therapeutic agents to bone is crucial in several diseases such as osteoporosis, Paget's disease, myeloproliferative diseases, multiple myeloma as well as skeletal metastasizing cancers. Prevention of cancer growth and lowering the cancer induced bone resorption is important in the treatment of bone metastasizing cancers. Keeping in mind the low diffusivity and availability of cell surface targets on cancer cells, we designed a targeted system to deliver chemotherapeutic agents to the bone microenvironment as an approach to tissue targeting using alendronate (Aln). We co-encapsulated curcumin and bortezomib in the PLGA nanoparticles to further enhance the therapeutic efficiency and overall clinical outcome. These multifunctional nanoparticles were characterized for particle size, morphology and drug encapsulation. The particles were spherical with smooth surface and had particle size of 235 ± 70.30 nm. We validated the bone targeting ability of these nanoparticles in vitro. Curcumin and bortezomib are known to have synergistic effect in inhibition of growth of cancer; however there was no synergism in the anti-osteoclastogenic activity of these agents. Surprisingly, curcumin by itself had significant inhibition of osteclastogenic activity. In vivo non-invasive bioimaging showed higher localization of Aln-coated nanoparticles to the bone compared to control groups, which was further confirmed by histological analysis. Aln-coated nanoparticles protected bone resorption and decreased the rate of tumor growth as compared to control groups in an intraosseous model of bone metastasis. Our data show efficient attachment of Aln on the surface of nanoparticles which could be used as a drug carrier for preferential delivery of multiple therapeutic agents to bone microenvironment.


Subject(s)
Alendronate/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/secondary , Bone and Bones , Boronic Acids/pharmacology , Bortezomib , Curcumin/pharmacology , Drug Carriers , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Osteoporosis , Polylactic Acid-Polyglycolic Acid Copolymer , Pyrazines/pharmacology
6.
J Cell Sci ; 124(Pt 9): 1453-64, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21486955

ABSTRACT

Extracellular proteolysis is an indispensable requirement for the formation of new blood vessels during neovascularization and is implicated in the generation of several angiogenic regulatory molecules. Anti-proteolytic agents have become attractive therapeutic strategies in diseases associated with excessive neovascularization. Annexin A2 (AnxA2) is an endothelial cell-surface receptor for the generation of active proteolytic factors, such as plasmin. Here, we show that AnxA2 is abundantly expressed in the neovascular tufts in a murine model of neovascularization. Exposure to hypoxic conditions results in elevation of AnxA2 and tissue plasminogen activator (tPA) in human retinal microvascular endothelial cells (RMVECs). We show that the hexapeptide competitive inhibitor LCKLSL, which targets the N-terminal tPA-binding site of AnxA2, binds efficiently to cell-surface AnxA2 compared with binding of the control peptide LGKLSL. Treatment with the competitive peptide inhibits the generation of plasmin and suppresses the VEGF-induced activity of tPA under hypoxic conditions. Application of the competitive peptide in two in vivo models of angiogenesis demonstrated suppression of the angiogenic responses, which was also associated with significant changes in the vascular sprouting. These results suggest that AnxA2-mediated plasmin generation is an important event in angiogenesis and is inhibited by a specific competitive peptide that inhibits the binding of tPA to AnxA2.


Subject(s)
Annexin A2/antagonists & inhibitors , Cell Hypoxia/physiology , Neovascularization, Pathologic/drug therapy , Oligopeptides/therapeutic use , Animals , Cells, Cultured , Chick Embryo , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Microscopy, Confocal , Neovascularization, Pathologic/chemically induced , Protein Binding/drug effects , Retina/cytology , Retina/drug effects , Retina/pathology , Tissue Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...