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.
ACS Appl Mater Interfaces ; 10(33): 27589-27602, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30048112

ABSTRACT

Targeting dendritic cells (DCs), either ex vivo (Ex. Sipuleucel-T) or in vivo, for stimulating cellular immunity has been a leading approach for cancer vaccines. We have rationally engineered a nanoparticle (NP)-based delivery system for vaccines (InAc-NPs) using inulin acetate (InAc) as the polymer to target DCs. The material and the antigen-encapsulated InAc-NPs (∼190 nm in diameter) were characterized for their physicochemical properties. As a potent vaccine adjuvant, InAc-NPs activated TLR4 on multiple immune cells, including DCs and primary swine and human cells, to secrete various cytokines as detected by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction. In addition, InAc-NPs promoted the maturation of DCs as observed by a decreased phagocytic ability and enhanced capability to activate various maturation markers (MHC-I, MHC-II, CD40, and CD80) quantified using flow cytometry. In mice, the InAc-NPs produced strong serum antibody titers (total IgG, IgG1, and IgG2a) against the encapsulated antigen (ovalbumin) similar to complete Freund's adjuvant. Additionally, as a dose-sparing delivery system, antigen delivered through InAc-NPs generated higher antibody titers (IgG1, 1.57 times; IgG-total, 1.66 times; and IgG2a, 29.8 times) even at 100 times less antigen dose. High amounts of cytokines representing both humoral (IL4 and IL10) and cell-mediated (IL2 and IFN-γ) immunities were secreted from splenocytes of mice immunized with InAc-NPs. Importantly, InAc-NPs provided complete protection in 100% of the vaccinated mice from metastasis of intravenously injected melanoma cells (B16-F10) to lungs. In addition, the InAc-NPs were cleared from the injection site within 30 h of injection (in vivo imaging) and displayed no toxicity at the injection site (histology). The current study demonstrates that the multifunctional InAc-based nanovaccine delivery system has potential applications in cancer immunotherapy and delivering vaccines against various infectious diseases.


Subject(s)
Dendritic Cells , Adjuvants, Immunologic , Animals , Antigens , Cancer Vaccines , Humans , Mice , Mice, Inbred C57BL , Nanoparticles , Polyglycolic Acid , Swine
2.
Int J Pharm ; 500(1-2): 32-41, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26792170

ABSTRACT

Micellization offers several advantages for the delivery of water insoluble drugs including a nanoparticulate 'core-shell' delivery system for drug targeting. Recently, hydrophobically modified polysaccharides (HMPs) are gaining recognition as micelle forming polymers to encapsulate hydrophobic drugs. In this manuscript, for the first time, we have evaluated the self-assembling properties of a lauryl carbamate derivative of the poly-fructose natural polymer inulin (Inutec SP1(®) (INT)) to form paclitaxel (PTX) loaded micelles. INT self-assembled into well-defined micellar structures in aqueous environment with a low critical micellar concentration of 27.8 µg/ml. INT micelles exhibited excellent hemocompatibility and low toxicity to cultured cells. PTX loaded INT micelles exhibited a mean size of 256.37 ± 10.45 nm with excellent drug encapsulation efficiency (95.66 ± 2.25%) and loading (8.69 ± 0.22%). PTX loaded micelles also displayed sustained release of PTX and enhanced anti-cancer efficacy in-vitro in mouse melanoma cells (B16F10) compared to Taxol formulation with Cremophor EL as solvent. In addition, PTX loaded INT micelles exhibited comparable in-vivo antitumor activity in B16F10 allograft mouse model at half the dose of Taxol. In conclusion, INT offers safe, inexpensive and natural alternative to widely used PEG-modified polymers for the formulation of micellar delivery systems for paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Carriers/administration & dosage , Inulin/administration & dosage , Paclitaxel/administration & dosage , Administration, Intravenous , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Drug Liberation , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Inulin/chemistry , Inulin/pharmacology , Inulin/therapeutic use , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Micelles , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Tumor Burden/drug effects
3.
Tumour Biol ; 37(2): 1727-38, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26314861

ABSTRACT

Epidemiological studies have demonstrated a significant correlation between regular aspirin use and reduced colon cancer incidence and mortality; however, the pathways by which it exerts its anti-cancer effects are still not fully explored. We hypothesized that aspirin's anti-cancer effect may occur through downregulation of c-Myc gene expression. Here, we demonstrate that aspirin and its primary metabolite, salicylic acid, decrease the c-Myc protein levels in human HCT-116 colon and in few other cancer cell lines. In total cell lysates, both drugs decreased the levels of c-Myc in a concentration-dependent fashion. Greater inhibition was observed in the nucleus than the cytoplasm, and immunofluorescence studies confirmed these observations. Pretreatment of cells with lactacystin, a proteasome inhibitor, partially prevented the downregulatory effect of both aspirin and salicylic acid, suggesting that 26S proteasomal pathway is involved. Both drugs failed to decrease exogenously expressed DDK-tagged c-Myc protein levels; however, under the same conditions, the endogenous c-Myc protein levels were downregulated. Northern blot analysis showed that both drugs caused a decrease in c-Myc mRNA levels in a concentration-dependent fashion. High-performance liquid chromatography (HPLC) analysis showed that aspirin taken up by cells was rapidly metabolized to salicylic acid, suggesting that aspirin's inhibitory effect on c-Myc may occur through formation of salicylic acid. Our result suggests that salicylic acid regulates c-Myc level at both transcriptional and post-transcription levels. Inhibition of c-Myc may represent an important pathway by which aspirin exerts its anti-cancer effect and decrease the occurrence of cancer in epithelial tissues.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/prevention & control , Proto-Oncogene Proteins c-myc/biosynthesis , Salicylates/pharmacology , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Chemoprevention , Chromatography, High Pressure Liquid , Down-Regulation , Fluorescent Antibody Technique , Gene Expression/drug effects , Humans , Neoplasms/pathology
4.
Nutr Cancer ; 67(3): 514-22, 2015.
Article in English | MEDLINE | ID: mdl-25751508

ABSTRACT

2-Deoxyglucose (2DG) is an anticancer drug with excellent safety profile. Because of its higher dose requirements, its potential is yet to translate into a monotherapy. However, recently, 2DG has been tested as an adjunct in established chemotherapeutic regimens. 2DG enhanced the potency of several chemotherapeutic agents but not all. The rationale selection of known chemotherapeutic agents to use with 2DG is hampered because of the lack of complete understanding of mechanism behind 2DG anticancer effects. Although, 2DG is a well-known glycolytic inhibitor, which inhibits the key glycolytic enzyme hexokinase, its anticancer effects cannot be fully explained by this simplistic mechanism alone. In this article, we have shown for the first time that 2DG induced a transient expression of p21 and a continuous expression of p53 in colorectal cancer cells (SW620). The treatment also caused cell cycle arrest at G0/G1 phase and induced apoptosis through the mitochondrial pathway. The effects of 2DG on p21 and p53 protein levels were totally independent of its inhibitory effect on either hexokinase or ATP levels. Results from this study provides key insights into novel molecular mechanisms of 2DG and directs rational selection of other anticancer drugs to combine with 2DG in colorectal cancer treatment.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Deoxyglucose/pharmacology , Glycolysis/physiology , Adenosine Triphosphate/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase Inhibitor p21/analysis , Humans , Tumor Suppressor Protein p53/analysis
5.
Mar Drugs ; 10(10): 2166-2180, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23170076

ABSTRACT

Sarcophine-diol (SD) is a semi-synthetic derivative of sarcophine with a significant chemopreventive effect against non-melanoma skin cancer both in vitro and in vivo. Recently, we have studied the effect of SD on melanoma development using the mouse melanoma B16F10 cell line. In this study, our findings show that SD suppresses cell multiplication and diminishes membrane permeability for ethidium bromide (EB), a model marker used to measure cell permeability for Ca²âº ions. SD also decreases protein levels of COX-2, and increases degradation of phospholipases PLA2 and PLC(γ)1 and diminishes enzymatic activity of the Ca²âº-dependent cPLA2. This lower membrane permeability for Ca²âº-ions, associated with SD, is most likely due to the diminished content of lysophosphosphatidylcholine (lysoPC) within cell membranes caused by the effect of SD on PLA2. The decrease in diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3) due to inhibition of PLC(γ)1, leads to the downregulation of Ca²âº-dependent processes within the cell and also inhibits the formation of tumors. These findings support our previous data suggesting that SD may have significant potential in the treatment of melanoma.


Subject(s)
Cell Membrane Permeability/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Diterpenes/pharmacology , Phospholipase C gamma/metabolism , Phospholipases A2, Cytosolic/antagonists & inhibitors , Animals , Anthozoa/chemistry , Anthozoa/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Cyclooxygenase 2/genetics , Diterpenes/chemistry , Diterpenes/metabolism , Gene Expression Regulation, Enzymologic , Melanoma/metabolism , Mice , Molecular Structure , Phospholipase C gamma/genetics , Phospholipases A2, Cytosolic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...