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










Publication year range
1.
ChemMedChem ; : e202400195, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687188

ABSTRACT

A series of N-glycosyl- 6BrCaQ conjugates was synthesized through a Pd-catalyzed cross-coupling reaction between brominated N-glycosyl quinolin-2-one derivatives and various nitrogen nucleophiles. Antiproliferative assays revealed that this new series of analogues represents a promising class of antitumor compounds as illustrated by the high biological activity observed for several derivatives towards different cancer cell lines compared to the non-glycosylated congeners.

2.
Chembiochem ; 25(10): e202400062, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38536125

ABSTRACT

This study evaluated the potential of isoCoQ-Carbazole, a diheterocyclic analog of isoCA-4, as an anti-tumor agent. To overcome its low aqueous solubility, liposomes were developed as a delivery system for the compound. In vitro experiments showed that loaded liposomes exhibited similar activity to the free form on multiple human tumor cell lines. In vivo experiments using a palliative intratumoral injection chemotherapy approach further demonstrated that isoCoQ-Carbazole loaded liposomes significantly reduced tumor growth in a CA-4-resistant HT29 cell model, without inducing any observable toxicity or weight loss in the treated mice. These findings suggest that liposomal isoCoQ-Carbazole may hold promise as a potential therapeutic agent for the treatment of inoperable, radiation-insensitive cancers.


Subject(s)
Antineoplastic Agents , Carbazoles , Liposomes , Solubility , Humans , Liposomes/chemistry , Carbazoles/chemistry , Carbazoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Animals , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Drug Screening Assays, Antitumor
3.
Int J Pharm ; 650: 123491, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-37806508

ABSTRACT

The development of novel cell-based therapies has increased the necessity to improve the long-term storage of cells. The current method of cryopreservation is far from optimal, causing ice-associated mechanical and osmotic damage to sensitive cells. Cell encapsulation is emerging as a new strategy to overcome those current limitations; however, few data are applicable to slow freezing, with conflicting results and multiple experimental conditions. The objective of this research work was to evaluate the impact of capsule size and encapsulation method on cell survival and functionality after a conventional freezing protocol. To this end, cells were encapsulated in alginate beads of different sizes, spanning the range of 200-2000 µm thanks to multiple extrusion techniques and conditions, and further cryopreserved using a slow cooling rate (-1°C/min) and 10 % DMSO as cryoprotectant. Our data show that there is a strong correlation between bead size and cell survival after a slow cooling cryopreservation process, with cell viabilities ranging from 7 to 70 % depending on the capsule size, with the smallest capsules (230 µm) achieving the highest level of survival. The obtained results indicate that the beads' diameter, rather than their morphology or the technique used, plays a significant role in the post-thawing cell survival and functionality. These results show that a fine control of cell encapsulation in alginate hydrogels is required when it comes to overcoming the current limitations of long-term preservation techniques by slow cooling.


Subject(s)
Dimethyl Sulfoxide , Hydrogels , Cell Survival , Cryopreservation/methods , Cryoprotective Agents/pharmacology , Alginates , Macrophages
4.
Biomacromolecules ; 24(2): 667-677, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36599673

ABSTRACT

Small interfering RNA (siRNA) holds promise for treating rheumatoid arthritis by inhibiting major cytokines such as tumor necrosis factor-α (TNF-α). We developed original cationic amphiphilic phosphorus dendrons to produce dendriplexes associated with TNF-α siRNA. The dendrons were made of 10 pyrrolidinium end groups and a C17 aliphatic chain. The dendriplexes demonstrated the ability to protect siRNA from nuclease degradation and to promote macrophage uptake. Moreover, they led to potent inhibition of TNF-α expression in the lipopolysaccharide-activated mouse macrophage cell line RAW264.7 in vitro model. A significant anti-inflammatory effect in the murine collagen-induced arthritis model was observed through arthritis scoring and histological observations. These results open up essential perspectives in using this original amphiphilic dendron to reduce the disease burden and improve outcomes in chronic inflammatory diseases.


Subject(s)
Arthritis, Experimental , Dendrimers , Animals , Mice , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Necrosis Factor-alpha/genetics , Anti-Inflammatory Agents/pharmacology
5.
Eur J Pharm Biopharm ; 170: 112-120, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34890789

ABSTRACT

In a strategy to improve macrophage targeting of glucocorticoids (GCs) for anti-inflammatory therapy, a so-called nanoprodrug of budesonide palmitate decorated by mannose moieties was designed. The synthesis of budesonide palmitate (BP) was obtained by esterification and mannosylated lipid (DSPE-PEG-Man) by reacting 1,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine (DSPE)-polyethylene glycol-amine and α-D-mannopyranosylphenyl isothiocyanate (MPITC). Nanoparticles were formulated by emulsion-evaporation and different ratios of mannosylated lipid were introduced in the formulation of BP nanoprodrugs. Using up to 75% of DSPE-PEG-man (75/25) led to 200 nm particles with a polydispersity index below 0.2, a negative zeta potential ranging from -10 to -30 mV, and one-month stability at 4 °C. The encapsulation efficiency of BP approached 100% proving that the prodrug was associated with the particles, leading to a final BP loading of 50-to 60% (w/w). The lectin agglutination test confirmed the availability of mannose on the nanoprodrug surface. Nanoprodrug uptake by RAW 264.7 macrophages was observed by confocal microscopy and flow cytometry. After 24 and 48 h of incubation, a significantly greater internalization of mannosylated nanoparticles as compared to PEGylated nanoparticles was achieved. The mannose receptor-mediated uptake was confirmed by a mannan inhibition study. After LPS-induced inflammation, the anti-inflammatory effect of mannosylated nanoparticles was assessed. After 48 h of incubation, cytokines (MCP-1 and TNFα) were reduced demonstrating that the functionalization of nanoprodrugs is possible and efficient.


Subject(s)
Budesonide/pharmacology , Mannose/pharmacology , Prodrugs/chemical synthesis , Animals , Biological Availability , Budesonide/administration & dosage , Cell Survival , Cells, Cultured , Cytokines/metabolism , Drug Compounding , Drug Delivery Systems , Drug Liberation , Drug Stability , Macrophages/drug effects , Mannose/administration & dosage , Mice , Nanoparticles , Tissue Distribution
6.
Int J Pharm ; 608: 121128, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34560204

ABSTRACT

Purified Glycogen (PG) is a highly hyper branched carbohydrate, characterized by high water solubility and very moderate increase in viscosity. The dendrimeric structure of PG, appropriately functionalized, makes it an alternative to current synthetic gene delivery agents. The present study explores the preparation of purified glycogen polycationic derivatives (PGPDs), developed and characterized starting from a single step reaction between PG and N,N-dialkylamino alkyl halides. Subsequently PGPDs were used for the complexation of a model siRNA nucleic acid, a transfection reagent siRNA and a fluorescein-labelled dsRNA oligomer. PGPDs-siRNA complexes were fully characterized by agarose gel electrophoresis and their efficacy was assessed by both confocal microscopy and transfection assays on breast and renal cancer cells. Results proved that PGPDs-siRNA complexes were efficient and not cytotoxic, maintaining their spherical and dendrimeric structure and, particularly, were able to effectively transfect the target cells by releasing the siRNA.


Subject(s)
Gene Transfer Techniques , Glycogen , Genetic Therapy , RNA, Small Interfering , Transfection
7.
Int J Pharm ; 598: 120381, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33610735

ABSTRACT

We propose to combine two therapeutic anti-inflammatory approaches with different mechanisms of action in a single drug delivery system consisting of cationic dexamethasone palmitate nanoparticles (CDXP-NP) associated with TNF-α siRNA. The CDXP-NPs are obtained by the solvent emulsion evaporation technique using dexamethasone palmitate, a prodrug of dexamethasone, associated with a cationic lipid, DOTAP. Their physicochemical properties as well as their ability to bind siRNA were evaluated through gel electrophoresis and siRNA binding quantification. SiRNA cellular uptake was assessed by flow cytometry and confocal microscopy on RAW264.7 macrophages. TNF-α inhibition was determined on LPS-activated RAW264.7 macrophages. Stable and monodisperse nanoparticles around 100 nm with a positive zeta potential (+59 mV) were obtained with an encapsulation efficiency of the prodrug of 95%. A nitrogen/phosphate (N/P) ratio of 10 was selected that conferred the total binding of siRNA to the nanoparticles. Using these CDXP-siRNA-NPs, the siRNA was strongly internalized by RAW264.7 macrophage cells and localized within the cytoplasm. On the LPS-induced RAW264.7 macrophages, a larger inhibition of TNF-α was observed with CDXP-siRNA-NPs compared to CDXP-NPs alone. In conclusion, from these data, it is clear that a combination of DXP and TNF-α siRNA therapy could be a novel strategy and optimized alternative approach to cure inflammatory diseases.


Subject(s)
Nanoparticles , Tumor Necrosis Factor-alpha , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , RNA, Small Interfering , Tumor Necrosis Factor-alpha/genetics
8.
Nanoscale ; 12(4): 2452-2463, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31915784

ABSTRACT

The understanding of the cellular uptake and the intracellular fate of nanoparticles and their subsequent influence on cell viability is challenging as far as micelles are concerned. Such systems are dynamic by nature, existing as unimers under their critical micelle concentration (CMC), and as micelles in equilibrium with unimers above the CMC, making canonical dose-response relationships difficult to establish. The purpose of this study was to investigate the in vitro cytotoxicity and uptake of two micellar sytems that are relevant for drug delivery. The two micelles incorporate a poly(ethylene glycol) coating and a pentacosadiynoic core which is either polymerized (pDA-PEG micelles) or non-polymerized (DA-PEG micelles), with the aim of evaluating the influence of the micelles status ("particle-like" or "dynamic", respectively) on their toxicological profile. Intracellular distribution and cytotoxicity of polymerized and non-polymerized micelles were investigated on RAW 264.7 macrophages in order to compare any different interactions with cells. Non-polymerized micelles showed significantly higher cytotoxicity than polymerized micelles, especially in terms of cell permeabilization, correlated to a higher accumulation in cell membranes. Other potential toxicity endpoints of polymerized micelles were then thoroughly studied in order to assess possible responses resulting from their endocytosis. No specific mechanisms of cytotoxicity were observed, neither in terms of apoptosis induction, cell membrane damage, release of inflammatory mediators nor genotoxicity. These data indicate that non-polymerized micelles accumulate in the cell membrane and induce cell membrane permeabilization, resulting in significant toxicity, whereas polymerized, stable micelles are internalized by cells but exert no or very low toxicity.


Subject(s)
Micelles , Polyacetylene Polymer/toxicity , Animals , Apoptosis , Drug Carriers , Endocytosis , Inflammation , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides , Mice , Mitochondria/metabolism , Nanoparticles/chemistry , Nanoparticles/toxicity , Nanostructures , Necrosis , Permeability , Polyacetylene Polymer/chemistry , Polyethylene Glycols/chemistry , Polymerization , RAW 264.7 Cells
9.
J Control Release ; 271: 98-106, 2018 02 10.
Article in English | MEDLINE | ID: mdl-29277682

ABSTRACT

In this study, we describe a liposome-based siRNA delivery system with a core composed of siRNA:protamine complex and a shell designed for the active targeting of CD44-expressing cells using for the first time the anti-CD44 aptamer (named Apt1) as targeting ligand. Among all functions, CD44 is the most common cancer stem cell surface biomarker and is found overexpressed in many tumors making this an attractive receptor for therapeutic targeting. This unique non-cationic system was evaluated for the silencing of the reporter gene of luciferase (luc2) in a triple-negative breast cancer model in vitro and in vivo. We show the possibility of conjugating an aptamer to siRNA-containing liposomes for an efficient gene silencing in CD44-expressing tumor cells in vivo, in the perspective of silencing disease-related genes in tumors.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Biomarkers, Tumor/genetics , Hyaluronan Receptors/genetics , RNA, Small Interfering/administration & dosage , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Gene Silencing , Humans , Liposomes , Luciferases/genetics , Mice , Nanomedicine , Triple Negative Breast Neoplasms/therapy
10.
Int J Pharm ; 514(1): 103-111, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27863652

ABSTRACT

We have investigated the impact of hyaluronic acid (HA)-coating on the targeting capacity of siRNA lipoplexes to CD44-overexpressing tumor cells. Cellular uptake and localization of HA-lipoplexes were evaluated by flow cytometry and fluorescence microscopy and both methods showed that these lipoplexes were rapidly internalized and localized primarily within the cytoplasm. Inhibition of luciferase expression on the A549-luciferase lung cancer cell line was achieved in vitro using an anti-Luc siRNA. 81% of luciferase gene expression inhibition was obtained in vitro with HA-lipoplexes at +/- ratio 2. In vivo, in a murine A549 metastatic lung cancer model, the treatment with HA-lipoplexes carrying anti-luciferase siRNA led to a statistically significant decrease of luciferase expression as opposed to progressive increase with non-modified lipoplexes or NaCl 0.9%. The reduction of the expression of luciferase mRNA tumor of mice treated with HA-lipoplexes supported the inhibition effect due to siRNA. These results highlight the potential of HA-lipoplexes in CD44-targeting siRNA delivery.


Subject(s)
Hyaluronic Acid/chemistry , Liposomes/chemistry , Lung Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Animals , Cell Line, Tumor , Female , Humans , Hyaluronan Receptors/metabolism , Luciferases/metabolism , Mice , RNA, Messenger/metabolism
11.
Biochim Biophys Acta ; 1858(11): 2725-2736, 2016 11.
Article in English | MEDLINE | ID: mdl-27457703

ABSTRACT

Phospholipid monolayers are often described as membrane models for analyzing drug-lipid interactions. In many works, a single phosphatidylcholine is chosen, sometimes with one or two additional components. Drug penetration is studied at 30mN/m, a surface pressure considered as corresponding to the pressure in bilayers, independently of the density of lipid molecular packing. In this work, we have extracted, identified, and quantified the major lipids constituting the lipidome of plasma and mitochondrial membranes of retinoblastoma (Y79) and retinal pigment epithelium cells (ARPE-19), using liquid chromatography coupled to high-resolution mass spectrometry (LC-MS/MS). The results obtained from this lipidomic analysis were used in an attempt to build an artificial lipid monolayer with a composition mimicking that of the plasma membrane of Y79 cells, better than a single phospholipid. The variety and number of lipid classes and species in cell extracts monolayers exceeding by far those of the phospholipids chosen to mimic them, the π-A isotherms of model monolayers differed from those of lipid extracts in shape and apparent packing density. We propose a model monolayer based on the most abundant species identified in the extracts, with a surface compressional modulus at 30mN/m close to the one of the lipid extracts.


Subject(s)
Cell Membrane/chemistry , Cholesterol/chemistry , Epithelial Cells/chemistry , Lipid Bilayers/chemistry , Phospholipids/chemistry , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/metabolism , Chromatography, Liquid , Epithelial Cells/metabolism , Humans , Lipid Bilayers/metabolism , Membranes, Artificial , Molecular Mimicry , Phospholipids/classification , Phospholipids/metabolism , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/metabolism , Tandem Mass Spectrometry
12.
Nanomedicine (Lond) ; 11(14): 1865-87, 2016 07.
Article in English | MEDLINE | ID: mdl-27389568

ABSTRACT

Extensive experimental evidence demonstrates the important role of hyaluronic acid (HA)-CD44 interaction in cell proliferation and migration, inflammation and tumor growth. Taking advantage of this interaction, the design of HA-modified nanocarriers has been investigated for targeting CD44-overexpressing cells with the purpose of delivering drugs to cancer or inflammatory cells. The effect of such modification on targeting efficacy is influenced by several factors. In this review, we focus on the impact of HA-modification on the characteristics of lipid-based nanoparticles. We try to understand how these modifications influence particle physicochemical properties, interaction with CD44 receptors, intracellular trafficking pathways, toxicity, complement/macrophage activation and pharmacokinetics. Our aim is to provide insight in tailoring particle modification by HA in order to design more efficient CD44-targeting lipid nanocarriers.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Hyaluronan Receptors/metabolism , Hyaluronic Acid/analogs & derivatives , Lipids/chemistry , Liposomes/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Humans , Hyaluronic Acid/metabolism , Lipid Metabolism , Liposomes/metabolism , Nanocapsules/chemistry , Neoplasms/metabolism
13.
Pharm Res ; 32(5): 1585-603, 2015 May.
Article in English | MEDLINE | ID: mdl-25416027

ABSTRACT

PURPOSE: We optimize the encapsulation and investigate the pharmacokinetics of 5-Fluorouracil (5-FU) delivered by thermosensitive stealth(®) liposomes (TSLs) designed to trigger drug release upon hyperthermia using focused ultrasound (FUS). METHODS: 5-FU was encapsulated into liposomes made of 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine/cholesterol/1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-PEG2000 either as a free molecule or complexed with copper-polyethylenimine. Heat-triggered drug release was evaluated using either a water bath or FUS. Formulation cytotoxicity was assessed on HT-29 cell line by MTS assay. Pharmacokinetics and biodistribution of 5-FU were evaluated in HT-29-tumor bearing mice. RESULTS: 5-FU was easily encapsulated using the lipid hydration method (encapsulation efficacy of 13%) but poorly retained upon dilution. 5-FU complexation with copper-polyethylenimine improved 5-FU retention into liposomes and allowed to obtain an encapsulation efficacy of 37%. At 42°C, heat-triggered 5-FU release from TSLs was 63% using a water bath and 68% using FUS, within 10 min, whereas it remained below 20% for the non-thermosensitive formulation. The MTS assay revealed that formulation toxicity arose from 5-FU and not from the excipients. In addition, 5-FU complex encapsulation into TSLs induces a reduction of the IC50 from 115 down to 49 µM. Pharmacokinetics reveals a longer circulation of encapsulated 5-FU and a more important body exposure, although tumor passive targeting is not significantly higher than free 5-FU. CONCLUSIONS: Complexation of 5-FU with copper-polyethylenimine appears an interesting strategy to improve 5-FU retention into TSLs in vitro and in vivo. TSLs allow heat-triggered release of the drug within 10 min at 42°C, a reasonable time for future in vivo experiments.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , 1,2-Dipalmitoylphosphatidylcholine/analogs & derivatives , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Animals , Antimetabolites, Antineoplastic/pharmacology , Cholesterol/chemistry , Drug Liberation , Fluorouracil/pharmacology , HT29 Cells , Humans , Hyperthermia, Induced , Liposomes/chemistry , Male , Mice , Mice, Inbred BALB C , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Tissue Distribution
14.
Int J Pharm ; 482(1-2): 75-83, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25448553

ABSTRACT

The purpose of this study was to investigate the toxicity of a series of poly(lactide-co-glycolic) (PLGA) nanoparticles on human-like THP-1 macrophages. Positively-, negatively-charged and neutral nanoparticles (200 nm) were prepared using chitosan (CS), poloxamer 188 (PF68) and poly(vinyl alcohol) (PVA) as stabilizer. Stabilizer-free PLGA nanoparticles were obtained as well. When used at therapeutically relevant concentrations (up to 0.1 mg/mL in vitro), all tested nanoparticles showed no or scarce signs of toxicity, as assessed by cell mitochondrial activity, induction of apoptosis and necrosis, production of intracellular reactive oxygen species (ROS) and secretion of pro-inflammatory cytokines. At high concentrations (above 1mg/mL), cytotoxicity was found to be induced by the presence of stabilizers, whatever the toxicological pattern of the stabilizer itself. While stabilizer-free PLGA nanoparticles exerted no cytotoxicity, the slightly cytotoxic CS polymer conferred PLGA nanoparticles significant cytotoxicity when used as nanoparticle stabilizer; more surprisingly, the otherwise innocuous PVA and PF68 polymers also conferred a significant cytotoxicity to PLGA nanoparticles. These results unveiled the critical toxicological contribution played by stabilizers used for the formulation of PLGA nanoparticles when used at high concentrations, which may have implications for local toxicities of PLGA-based nanomedicine, and provided additional insight in cytotoxic effects of internalized nanoparticles.


Subject(s)
Excipients/toxicity , Lactic Acid/toxicity , Macrophages/drug effects , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyglycolic Acid/toxicity , Cell Survival/drug effects , Chitosan/chemistry , Chitosan/toxicity , Excipients/chemistry , Humans , Lactic Acid/chemistry , Poloxamer/chemistry , Poloxamer/toxicity , Polyglycolic Acid/chemistry , Polyvinyl Alcohol/chemistry , Polyvinyl Alcohol/toxicity
15.
Bioconjug Chem ; 26(7): 1307-13, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25343502

ABSTRACT

CD44 receptor protein is found to be overexpressed by many tumors and is identified as one of the most common cancer stem cell surface markers including tumors affecting colon, breast, pancreas, and head and neck, making this an attractive receptor for therapeutic targeting. In this study, 2'-F-pyrimidine-containing RNA aptamer (Apt1), previously selected against CD44, was successfully conjugated to the surface of PEGylated liposomes using the thiol-maleimide click reaction. The conjugation of Apt1 to the surface of liposomes was confirmed by the change in size and zeta potential and by migration on agarose gel electrophoresis. The binding affinity of Apt1 was improved after conjugation compared to free-Apt1. The cellular uptake for Apt1-Lip was tested by flow cytometry and confocal imaging using the two CD44(+) cell lines, human lung cancer cells (A549) and human breast cancer cells (MDA-MB-231), and the CD44(-) cell line, mouse embryonic fibroblast cells (NIH/3T3). The results showed higher sensitivity and selectivity for Apt1-Lip compared to the blank liposomes (Mal-Lip). In conclusion, we demonstrate a successful conjugation of anti-CD44 aptamer to the surface of liposome and binding preference of Apt1-Lip to CD44-expressing cancer cells and conclude to a promising potency of Apt1-Lip as a specific drug delivery system.


Subject(s)
Aptamers, Nucleotide/metabolism , Hyaluronan Receptors/metabolism , Liposomes/metabolism , Neoplasms/metabolism , Animals , Aptamers, Nucleotide/chemistry , Cell Line, Tumor , Drug Delivery Systems , Humans , Liposomes/chemistry , Mice , NIH 3T3 Cells , Neoplasms/drug therapy
16.
Bioconjug Chem ; 25(11): 1971-83, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25313527

ABSTRACT

We report herein a detailed study concerning the impact of different bioconjugation and nanoformulation strategies on the in vitro targeting ability of peptide-decorated squalenoyl gemcitabine (SQdFdC) nanoparticles (NPs). NPs have been functionalized with the CKAAKN peptide, previously identified as an efficient homing device within the pancreatic pathological microenvironment. Two approaches have been followed: (i) either the CKAAKN peptide was directly conjugated at the surface of preformed SQdFdC nanoparticles (conjugation after NP formation) or (ii) it was first reacted with a maleimide squalenoyl derivative before the resulting bioconjugate was co-nanoprecipitated with SQdFdC to form the peptide-decorated NPs (conjugation before NP formation). NPs were characterized with respect to mean diameter, zeta potential, and stability over time. Then, their specific interaction with the sFRP-4 protein was evaluated by surface plasmon resonance. Although both synthetic strategies allowed us to formulate NPs able to interact with the corresponding receptor, enhanced target binding and better specific avidity were observed with CKAAKN-NPs functionalized before NP formation. These NPs displayed the highest cell uptake and cytotoxicity in an in vitro model of human MIA Paca-2 pancreatic cancer cells.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Oligopeptides/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Drug Carriers/metabolism , Drug Design , Humans , Ligands , Mice , NIH 3T3 Cells , Nanoparticles/metabolism , Particle Size , Proto-Oncogene Proteins/metabolism , Gemcitabine
17.
J Control Release ; 192: 29-39, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-24984010

ABSTRACT

Chemotherapy for pancreatic cancer is hampered by the tumor's physio-pathological complexity. Here we show a targeted nanomedicine using a new ligand, the CKAAKN peptide, which had been identified by phage display, as an efficient homing device within the pancreatic pathological microenvironment. Taking advantage of the squalenoylation platform, the CKAAKN peptide was conjugated to squalene (SQCKAAKN) and then co-nanoprecipitated with the squalenoyl prodrug of gemcitabine (SQdFdC) giving near monodisperse nanoparticles (NPs) for safe intravenous injection. By interacting with a novel target pathway, the Wnt-2, the CKAAKN functionalization enabled nanoparticles: (i) to specifically interact with both tumor cells and angiogenic vessels and (ii) to simultaneously promote pericyte coverage, thus leading to the normalization of the vasculature likely improving the tumor accessibility for therapy. All together, this approach represents a unique targeted nanoparticle design with remarkable selectivity towards pancreatic cancer and multiple mechanisms of action.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Peptides/chemistry , Prodrugs/administration & dosage , Squalene/chemistry , Amino Acid Sequence , Animals , Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , Humans , Male , Mice, Inbred C57BL , Nanoparticles/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides/metabolism , Prodrugs/therapeutic use , Squalene/metabolism , Gemcitabine
18.
J Biomed Nanotechnol ; 10(10): 2852-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25992421

ABSTRACT

Biodegradable nanoparticles exhibit high potentialities for local or systemic drug delivery through lung administration making them attractive as nanomedicine carriers. However, since particulate matter or some inorganic manufactured nanoparticles exposed to lung cells have provoked cytotoxic effects, inflammatory and oxidative stress responses, it becomes important to investigate nanomedicine toxicity towards the lungs. This is the reason why, in the present review, the behavior of biodegradable nanoparticles towards the different parts of the respiratory tract as well as the toxicological consequences, measured on several models in vitro, ex vivo or in vivo, are described. Taken all together, the different studies carried out so far conclude on no or slight toxicity of biodegradable nanoparticles.


Subject(s)
Biocompatible Materials/toxicity , Lung/drug effects , Lung/pathology , Nanoparticles/toxicity , Animals , Coculture Techniques , Disease Models, Animal , Humans , Toxicity Tests
19.
Int J Pharm ; 454(2): 686-94, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23747506

ABSTRACT

In vitro cytotoxicity and inflammatory response following exposure to nanoparticles (NPs) made of poly(lactide-co-glycolide) (PLGA) have been investigated on A549 human lung epithelial cells. Three different PLGA NPs (230 nm) were obtained using different stabilizers (polyvinyl alcohol, chitosan, or Pluronic(®) F68) to form respectively neutral, positively or negatively charged NPs. Polystyrene NPs were used as polymeric but non-biodegradable NPs, and titanium dioxide (anatase and rutile) as inorganic NPs, for comparison. Cytotoxicity was evaluated through mitochondrial activity as well as membrane integrity (lactate dehydrogenase release, trypan blue exclusion, propidium iodide staining). The cytotoxicity of PLGA-based and polystyrene NPs was lower or equivalent to the one observed after exposure to titanium dioxide NPs. The inflammatory response, evaluated through the release of the IL-6, IL-8, MCP-1, TNF-α cytokines, was low for all NPs. However, some differences were observed, especially for negative PLGA NPs that led to a higher inflammatory response, which can be correlated to a higher uptake of these NPs. Taken together, these results show that both coating of PLGA NPs and the nature of the core play a key role in cell response.


Subject(s)
Epithelial Cells/drug effects , Lactic Acid/toxicity , Nanoparticles/toxicity , Polyglycolic Acid/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Chitosan/chemistry , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Lactic Acid/chemistry , Nanoparticles/chemistry , Poloxamer/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polyvinyl Alcohol/chemistry , Pulmonary Alveoli/cytology , Surface Properties
20.
Int J Pharm ; 454(2): 703-11, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23701998

ABSTRACT

The aim of these studies was to synthesize, characterize and evaluate the efficacy of pegylated gold nanoparticles (AuNPs) that differed in their PEG molecular weight, using PEG 550 and PEG 2000. The synthesis of the gold nanoparticles was carried out by modified Brust method with a diameter of 4-15 nm. The targeting agent folic acid was introduced by the covalent linkage. Finally, the anti-cancer drug docetaxel was encapsulated by the AuNPs by non covalent adsorption. The nanoparticles were characterized by transmission electron microscopy and used for in vitro studies against a hormone-responsive prostate cancer cell line, LnCaP. The loaded nanoparticles reduced the cell viability in more than 50% at concentrations of 6 nM and above after 144 h of treatment. Moreover, observation of prostate cancer cells by optical microscopy showed damage to the cells after exposure to drug-loaded AuNPs while unloaded AuNPs had much less effect.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Taxoids/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Drug Carriers/administration & dosage , Folic Acid/chemistry , Gold/administration & dosage , Humans , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Polyethylene Glycols/chemistry , Taxoids/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...