Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomaterials ; 32(17): 4118-29, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21429574

ABSTRACT

There is an urgent need for novel polymeric carriers that can selectively deliver a large dose of chemotherapeutic agents into hepatic cancer cells to achieve high therapeutic activity with minimal systemic side effects. PAMAM dendrimers are characterized by a unique branching architecture and a large number of chemical surface groups suitable for coupling of chemotherapeutic agents. In this article, we report the coupling of N-acetylgalactosamine (NAcGal) to generation 5 (G5) of poly(amidoamine) (PAMAM-NH2) dendrimers via peptide and thiourea linkages to prepare NAcGal-targeted carriers used for targeted delivery of chemotherapeutic agents into hepatic cancer cells. We describe the uptake of NAcGal-targeted and non-targeted G5 dendrimers into hepatic cancer cells (HepG2) as a function of G5 concentration and incubation time. We examine the contribution of the asialoglycoprotein receptor (ASGPR) to the internalization of NAcGal-targeted dendrimers into hepatic cancer cells through a competitive inhibition assay. Our results show that uptake of NAcGal-targeted G5 dendrimers into hepatic cancer cells occurs via ASGPR-mediated endocytosis. Internalization of these targeted carriers increased with the increase in G5 concentration and incubation time following Michaelis-Menten kinetics characteristic of receptor-mediated endocytosis. These results collectively indicate that G5-NAcGal conjugates function as targeted carriers for selective delivery of chemotherapeutic agents into hepatic cancer cells.


Subject(s)
Acetylgalactosamine/pharmacokinetics , Antineoplastic Agents/pharmacology , Dendrimers/chemical synthesis , Dendrimers/pharmacokinetics , Drug Carriers/chemical synthesis , Polyamines/pharmacology , Biological Transport , Drug Delivery Systems/methods , Endocytosis , Hep G2 Cells , Humans , Polyamines/chemistry , Polymers
2.
Mol Pharm ; 7(6): 2006-19, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-20957996

ABSTRACT

Effective treatment of solid tumors requires homogeneous distribution of anticancer drugs within the entire tumor volume to deliver lethal concentrations to resistant cancer cells and tumor-initiating cancer stem cells. However, penetration of small molecular weight chemotherapeutic agents and drug-loaded polymeric and lipid particles into the hypoxic and necrotic regions of solid tumors remains a significant challenge. This article reports the results of pulsed ultrasound enhanced penetration of nanosized fluorescent particles into MCF-7 breast cancer spheroids (300-350 µm diameter) as a function of particle size and charge. With pulsed ultrasound application in the presence of microbubbles, small (20 nm) particles achieve 6-20-fold higher penetration and concentration in the spheroid's core compared to those not exposed to ultrasound. Increase in particle size to 40 and 100 nm results in their effective penetration into the spheroid's core to 9- and 3-fold, respectively. In addition, anionic carboxylate particles achieved higher penetration (2.3-, 3.7-, and 4.7-fold) into the core of MCF-7 breast cancer spheroids compared to neutral (2.2-, 1.9-, and 2.4-fold) and cationic particles (1.5-, 1.4-, and 1.9-fold) upon US exposure for 30, 60, and 90 s under the same experimental conditions. These results demonstrate the feasibility of utilizing pulsed ultrasound to increase the penetration of nanosized particles into MCF-7 spheroids mimicking tumor tissue. The effects of particle properties on the penetration enhancement were also illustrated.


Subject(s)
Breast Neoplasms/chemistry , Drug Delivery Systems , Microbubbles , Nanoparticles/chemistry , Spheroids, Cellular/chemistry , Ultrasonic Therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Fluorescence , Humans , Models, Biological , Particle Size , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Surface Properties , Tumor Cells, Cultured
3.
Ultrasound Med Biol ; 36(7): 1176-87, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20620704

ABSTRACT

Localized delivery of therapeutic agents through the blood-brain barrier (BBB) is a clinically significant task that remains challenging. Ultrasound (US) application after intravenous administration of microbubbles has been shown to generate localized BBB opening in animal models but the detailed mechanisms are not yet fully described. The current study investigates the effects of US-stimulated microbubbles on in vitro murine brain microvascular endothelial (bEnd.3) cells by monitoring sonoporation and changes in intracellular calcium concentration ([Ca(2+)](i)) using real-time fluorescence and high-speed brightfield microscopy. Cells seeded in microchannels were exposed to a single US pulse (1.25 MHz, 10 cycles, 0.24 MPa peak negative pressure) in the presence of Definity microbubbles and extracellular calcium concentration [Ca(2+)](o) = 0.9 mM. Disruption of the cell membrane was assessed using propidium iodide (PI) and change in the [Ca(2+)](i) was measured using fura-2. Cells adjacent to a microbubble exhibited immediate [Ca(2+)](i) changes after US pulse with and without PI uptake and the [Ca(2+)](i) changes were twice as large in cells with PI uptake. Cell viability assays showed that sonoporated cells could survive with modulation of [Ca(2+)](i) and uptake of PI. Cells located near sonoporated cells were observed to exhibit changes in [Ca(2+)](i) that were delayed from the time of US application and without PI uptake. These results demonstrate that US-stimulated microbubbles not only directly cause changes in [Ca(2+)](i) in brain endothelial cells in addition to sonoporation but also generate [Ca(2+)](i) transients in cells not directly interacting with microbubbles, thereby affecting cells in larger regions beyond the cells in contact with microbubbles.


Subject(s)
Calcium/metabolism , Cerebrovascular Circulation/physiology , Endothelial Cells/physiology , Microcirculation/physiology , Signal Transduction/physiology , Sonication , Animals , Cell Line , Cerebrovascular Circulation/radiation effects , Endothelial Cells/radiation effects , Mice , Microcirculation/radiation effects , Radiation Dosage , Signal Transduction/drug effects
4.
Nanoscale ; 2(5): 755-62, 2010 May.
Article in English | MEDLINE | ID: mdl-20648321

ABSTRACT

Poly(amidoamine) (PAMAM) dendrimers are a family of water-soluble polymers with a characteristic tree-like branching architecture and a large number of surface groups, which have been used to immobilize a variety of therapeutic molecules for targeted drug delivery. Earlier studies showed that small cationic PAMAM-NH2 and selected anionic PAMAM-COOH dendrimers permeate across in vitro models of the small intestinal epithelium by paracellular and transcellular transport mechanisms. The focus of this research is to mathematically calculate the effect of cationic, anionic, and neutral PAMAM dendrimers on the porosity of epithelial tight junctions as a function of dendrimers concentration, incubation time, generation number, and charge density. Results show that the increase in the concentration, incubation time and generation number of cationic G0-G2 PAMAM-NH2 and anionic G2.5 and G3.5 PAMAM-COOH dendrimers caused a corresponding increase in the porosity of Caco-2 cell monolayers. Neutral G2-G4 PAMAM-OH dendrimers had no effect on the porosity of intestinal cells. These results provide quantitative evidence that the observed increase in permeability of PAMAM dendrimers across Caco-2 cell monolayers is due to their effect on the organization of the tight junctions and the associated increase in membrane porosity. Furthermore, these results emphasize the potential of cationic PAMAM-NH2 and anionic PAMAM-COOH dendrimers to function as carriers for controlled oral drug delivery.


Subject(s)
Dendrimers/chemistry , Caco-2 Cells , Cell Membrane Permeability , Drug Carriers/chemistry , Humans , Mannitol/administration & dosage , Porosity
5.
Biomaterials ; 31(27): 7150-66, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20579726

ABSTRACT

This report describes the design and synthesis of a new series of degradable, pH-sensitive, membrane-destabilizing, comb-like polymers that can enhance the intracellular delivery of therapeutic nucleic acids. These comb-like polymers are based on a diblock polymer backbone where the first block is a copolymer of pH-sensitive ethyl acrylic acid (EAA) monomers and hydrophobic butyl methacrylate (BMA) or hexyl methacrylate monomers. The second block is a homopolymer of N-acryloxy succinimide (NASI) or ss-benzyl l-aspartate N-carboxy-anhydride (BLA-NCA) monomers, which are functionalized to allow controlled grafting of hydrophobic HMA and cationic trimethyl aminoethyl methacrylate (TMAEMA) copolymers via acid-labile hydrazone linkages. These comb-like polymers displayed high hemolytic activity in acidic solutions, which increased with the increase in polymer concentration. All comb-like polymers degraded into small fragments upon incubation in an acidic solution (pH 5.8) due to hydrolysis of the hydrazone linkages connecting the hydrophobic/cationic grafts to the polymer backbone. Comb-like polymers successfully complexed anti-GAPDH siRNA molecules into serum- and nuclease-stable particles, which successfully silenced GAPDH expression at both the mRNA and protein levels. These results collectively indicate the potential of these new comb-like polymers to serve as vehicles for effective intracellular delivery of therapeutic nucleic acids.


Subject(s)
Genetic Vectors/metabolism , Nucleic Acids/administration & dosage , Polymers/metabolism , Transfection/methods , Cell Line, Tumor , Drug Stability , Genetic Vectors/chemical synthesis , Genetic Vectors/chemistry , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Nucleic Acids/chemistry , Nucleic Acids/genetics , Polymers/chemical synthesis , Polymers/chemistry
6.
Anal Chem ; 82(6): 2505-11, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20178370

ABSTRACT

This manuscript describes a straightforward fabrication process for embedding Ag/AgCl electrodes within a two-layer poly(dimethylsiloxane) (PDMS) microfluidic chip where an upper and a lower channel are separated by a semiporous membrane. This system allows for the reliable real-time measurement of transendothelial and transepithelial electrical resistance (TEER), an accepted quantification of cell monolayer integrity, across cells cultured on membranes inside the microchannels using impedance spectroscopy. The technique eliminates the need for costly or specialized microelectrode fabrication, enabling commercially available wire electrodes to easily be incorporated into PDMS microsystems for measuring TEER under microfluidic environments. The capability of measuring impedance across a confluent cell monolayer is confirmed using (i) brain-derived endothelial cells (bEND.3), (ii) Madin Darby Canine Kidney Cells (MDCK-2), and mouse myoblast (C2C12) (all from ATCC, Manassas, VA). TEER values as a function of cell type and cell culture time were measured and both agree with previously published values from macroscale culture techniques. This system opens new opportunities for conveniently resolving both transendothelial and transepithelial electrical resistance to monitor cell function in real-time in microfluidic cell cultures.


Subject(s)
Electric Impedance , Electrochemistry/instrumentation , Endothelium/metabolism , Epithelial Cells/metabolism , Microfluidic Analytical Techniques/instrumentation , Animals , Cell Line , Dimethylpolysiloxanes/chemistry , Electrodes , Equipment Design , Membranes, Artificial
8.
J Biomater Sci Polym Ed ; 19(10): 1333-46, 2008.
Article in English | MEDLINE | ID: mdl-18854126

ABSTRACT

An excessive inflammatory response is a clinical problem following major infections and severe injury that may lead to Sepsis Syndrome and Multiple Organ Failure (MOF), including the Acute Respiratory Distress Syndrome (ARDS). Management of excessive inflammation may be possible through control of key inflammatory pathways such as those mediated by the important interleukin-1 receptor associated kinase-1 (IRAK-1). In the current study, we report the impact on gene expression induced by lipopolysaccharide (LPS) stimulation of THP-1 cells treated with an antisense oligonucleotide (ASODN) against the IRAK-1 gene using cDNA microarrays and quantitative RT-PCR. The therapeutic ASODN was delivered using a pH-sensitive, membrane-interactive polymer that destabilizes the endosomal membrane to enhance access cytoplasmic delivery in targeted cells. Following LPS stimulation, the anti-inflammatory activity of ASODN against the IRAK-1 gene expression is evidenced by the lower expression of inflammatory chemokines, cytokines and acute-phase proteins compared to control cells. These results provide a larger mechanistic picture of IRAK-1 knockdown by this polymer therapeutic in macrophage-like cells.


Subject(s)
Gene Knockdown Techniques , Interleukin-1 Receptor-Associated Kinases/genetics , Macrophages/drug effects , Macrophages/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Polymers/metabolism , Animals , Base Sequence , Cattle , Cell Line, Tumor , Chemokines/genetics , Chemokines/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA, Complementary/genetics , Electrophoretic Mobility Shift Assay , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Interleukins/genetics , Interleukins/metabolism , Lipopolysaccharides/genetics , Lipopolysaccharides/metabolism , Macrophages/cytology , Metallothionein/genetics , Metallothionein/metabolism , NF-kappa B/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Oligonucleotide Array Sequence Analysis , Polymers/chemical synthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
9.
Biomacromolecules ; 7(8): 2407-14, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16903689

ABSTRACT

Many macromolecular therapeutics such as peptides, proteins, antisense oligodeoxynucleotides (ASODN), and short interfering RNA (siRNA) are active only in the cytoplasm or nucleus of targeted cells. Endocytosis is the primary route for cellular uptake of these molecules, which results in their accumulation in the endosomal-lysosomal trafficking pathway and loss of therapeutic activity. In this article, we describe the synthesis and pH-dependent membrane-destabilizing activity of a new "smart" polymer family that can be utilized to enhance the intracellular delivery of therapeutic macromolecules through the endosomal membrane barrier into the cytoplasm of targeted cells. These polymers are propylamine, butylamine, and pentylamine derivatives of poly(styrene-alt-maleic anhydride) (PSMA) copolymers. The PSMA-alkylamide derivatives are hydrophilic and membrane-inactive at physiological pH; however, they become hydrophobic and membrane-disruptive in response to endosomal pH values as measured by their hemolytic activity. Results show that the pH-dependent membrane-destabilizing activity of PSMA derivatives can be controlled by varying the length of the alkylamine group, the degree of modification of the copolymer, and the molecular weight of the PSMA copolymer backbone. Butylamine and pentylamine derivatives of PSMA copolymers exhibited more than 80% hemolysis at endosomal pH values, which suggests their potential as a platform of "smart" polymeric carriers for enhanced cytoplasmic delivery of a variety of therapeutic macromolecules.


Subject(s)
Drug Delivery Systems , Maleic Anhydrides/chemical synthesis , Polymers/chemical synthesis , Polystyrenes/chemical synthesis , Animals , Endosomes/chemistry , Erythrocytes/chemistry , Erythrocytes/cytology , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Maleic Anhydrides/chemistry , Maleic Anhydrides/pharmacokinetics , Mice , NIH 3T3 Cells , Polymers/chemistry , Polymers/pharmacokinetics , Polystyrenes/chemistry , Polystyrenes/pharmacology
10.
Adv Drug Deliv Rev ; 57(15): 2163-76, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16289433

ABSTRACT

This article summarizes our efforts to evaluate the potential of poly (amidoamine) (PAMAM) dendrimers as carriers for oral drug delivery. Specifically, the permeability of a series of cationic PAMAM-NH2 (G0-G4) dendrimers across Caco-2 cell monolayers was evaluated as a function of dendrimer generation, concentration, and incubation time. The influence of dendrimer surface charge on the integrity, paracellular permeability, and viability of Caco-2 cell monolayers was monitored by measuring the transepithelial electrical resistance (TEER), 14C-mannitol permeability, and leakage of lactate dehydrogenase (LDH) enzyme, respectively. Microvascular extravasation of PAMAM-NH2 dendrimers in relation to their size, molecular weight, and molecular geometry is also discussed. Results of these studies show that transepithelial transport and microvascular extravasation of PAMAM dendrimers are dependent on their structural features including molecular size, molecular geometry, and surface chemistry. These results suggest that by optimizing the size and surface charge of PAMAM dendrimers, it is possible to develop oral delivery systems based on these carriers for targeted drug delivery.


Subject(s)
Dendrimers/pharmacokinetics , Polyamines/pharmacokinetics , Animals , Biological Transport, Active , Capillaries/metabolism , Drug Carriers , Drug Delivery Systems , Epithelium/metabolism , Humans
11.
J Control Release ; 104(2): 417-27, 2005 May 18.
Article in English | MEDLINE | ID: mdl-15984055

ABSTRACT

Limited cytoplasmic delivery of enzyme-susceptible drugs remains a significant challenge facing the development of protein and nucleic acid therapies that act in intracellular compartments. "Smart" pH-responsive, membrane-destabilizing polymers present a new approach to shuttling therapeutic molecules past the endosomal membrane and into the cytoplasm of targeted cells. This report describes the use of a functionalized monomer, pyridyl disulfide acrylate (PDSA), to develop pH-responsive, membrane-destabilizing, and glutathione-reactive polymers by copolymerization with several pH-responsive and hydrophobic monomers. The activity of the carriers is described as a function of (a) increasing the length of the hydrophobic alkyl group substituted onto the pH-responsive monomer and (b) the incorporation of a hydrophobic monomer such as butyl acrylate (BA) on the pH sensitivity and membrane-destabilizing activity of new polymer compositions. The membrane-destabilizing activity of different polymer compositions was evaluated as a function of pH and polymer concentration using the red blood cell (RBC) hemolysis assay. Hemolysis results show that the increase in the hydrophobic character of the polymer backbone results in a shift in the pH sensitivities and an increase in the membrane-destabilizing activity. Results show that the observed hemolytic activities and pH sensitivity profiles could be designed across a range that matches the properties needed for enhancing the cytoplasmic delivery of macromolecular therapeutic.


Subject(s)
Acrylates/chemical synthesis , Disulfides/chemical synthesis , Drug Design , Glutathione/metabolism , Polymers/chemical synthesis , Acrylates/pharmacology , Cell Membrane/drug effects , Disulfides/pharmacology , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Polymers/pharmacology
12.
Expert Opin Biol Ther ; 5(1): 23-32, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15709907

ABSTRACT

Limited cytoplasmic delivery of enzyme-susceptible drugs remains a significant challenge facing the development of protein and nucleic acid therapies that act in intracellular compartments. Researchers have examined several approaches, including fusogenic proteins and protein transduction domains, to enhance the intracellular delivery of the therapeutic cargo. This review summarises efforts to develop 'smart' pH-sensitive and membrane-destabilising polymers that can shuttle therapeutic peptide, protein and nucleic acid molecules past the endosomal membrane into the cytoplasm of targeted cells. Several classes of 'smart' non-degradable polymeric carriers have been developed that have proved effective both in vitro and in vivo in enhancing the cytoplasmic delivery of a variety of therapeutic molecules.


Subject(s)
Drug Carriers/administration & dosage , Intracellular Fluid/drug effects , Macromolecular Substances/administration & dosage , Polymers/administration & dosage , Animals , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Humans , Intracellular Fluid/metabolism , Macromolecular Substances/pharmacokinetics , Polymers/pharmacokinetics
13.
J Control Release ; 101(1-3): 47-58, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15588893

ABSTRACT

Limited cytoplasmic delivery of enzyme-susceptible drugs remains a significant challenge facing the development of protein and nucleic acid therapies that act in intracellular compartments. "Smart" pH-sensitive, membrane-destabilizing polymers present an attractive approach to shuttle therapeutic molecules past the endosomal membrane and into the cytoplasm of targeted cells. This report describes the use of a new functionalized monomer, pyridyl disulfide acrylate (PDSA), to develop pH-sensitive, membrane-destabilizing, and glutathione-reactive polymers by copolymerization with several pH-sensitive and hydrophobic monomers. The activity of the carriers is described as a function of (a) the influence of increasing the length of the hydrophobic alkyl group substituted onto the pH-sensitive monomer and (b) of the effect of incorporating a hydrophobic monomer such as butyl acrylate (BA) on the pH sensitivity and membrane-destabilizing activity of new polymer compositions. The membrane-destabilizing activity of different polymer compositions was evaluated as a function of pH and polymer concentration using the red blood cells (RBC) hemolysis assay. Hemolysis results show that the increase in the hydrophobic character of polymer backbone results in a shift in the pH sensitivity profile and an increase in the membrane-destabilizing activity. Results show that the observed hemolytic activities and pH sensitivity profiles could be designed across a range that matches the properties needed for drug carriers to enhance the cytoplasmic delivery of therapeutic cargos.


Subject(s)
Acrylates/chemical synthesis , Disulfides/chemical synthesis , Drug Delivery Systems , Drug Design , Glutathione/metabolism , Polymers/chemical synthesis , Acrylates/pharmacology , Cell Membrane/drug effects , Disulfides/pharmacology , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Molecular Weight , Polymers/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...