Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 4: e903, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24201802

ABSTRACT

Several neurodegenerative diseases and brain injury involve reactive oxygen species and implicate oxidative stress in disease mechanisms. Hydrogen peroxide (H2O2) formation due to mitochondrial superoxide leakage perpetuates oxidative stress in neuronal injury. Catalase, an H2O2-degrading enzyme, thus remains an important antioxidant therapy target. However, catalase therapy is restricted by its labile nature and inadequate delivery. Here, a nanotechnology approach was evaluated using catalase-loaded, poly(lactic co-glycolic acid) nanoparticles (NPs) in human neuronal protection against oxidative damage. This study showed highly efficient catalase encapsulation capable of retaining ~99% enzymatic activity. NPs released catalase rapidly, and antioxidant activity was sustained for over a month. NP uptake in human neurons was rapid and nontoxic. Although human neurons were highly sensitive to H2O2, NP-mediated catalase delivery successfully protected cultured neurons from H2O2-induced oxidative stress. Catalase-loaded NPs significantly reduced H2O2-induced protein oxidation, DNA damage, mitochondrial membrane transition pore opening and loss of cell membrane integrity and restored neuronal morphology, neurite network and microtubule-associated protein-2 levels. Further, catalase-loaded NPs improved neuronal recovery from H2O2 pre-exposure better than free catalase, suggesting possible applications in ameliorating stroke-relevant oxidative stress. Brain targeting of catalase-loaded NPs may find wide therapeutic applications for oxidative stress-associated acute and chronic neurodegenerative disorders.


Subject(s)
Catalase/metabolism , Nanoparticles/chemistry , Neurons/drug effects , Neurons/metabolism , Catalase/administration & dosage , Cells, Cultured , Humans , Hydrogen Peroxide/pharmacology , Lactic Acid/chemistry , Oxidative Stress/drug effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
2.
Cancer Gene Ther ; 19(8): 530-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22595792

ABSTRACT

Mutation of the p53 tumor suppressor gene, the most common genetic alteration in human cancers, results in more aggressive disease and increased resistance to conventional therapies. Aggressiveness may be related to the increased angiogenic activity of cancer cells containing mutant p53. To restore wild-type p53 function in cancer cells, we developed polymeric nanoparticles (NPs) for p53 gene delivery. Previous in vitro and in vivo studies demonstrated the ability of these NPs to provide sustained intracellular release of DNA, thus sustained gene transfection and decreased tumor cell proliferation. We investigated in vivo mechanisms involved in NP-mediated p53 tumor inhibition, with focus on angiogenesis. We hypothesize that sustained p53 gene delivery will help decrease tumor angiogenic activity and thus reduce tumor growth and improve animal survival. Xenografts of p53 mutant tumors were treated with a single intratumoral injection of p53 gene-loaded NPs (p53NPs). We observed intratumoral p53 gene expression corresponding to tumor growth inhibition, over 5 weeks. Treated tumors showed upregulation of thrombospondin-1, a potent antiangiogenic factor, and a decrease in microvessel density vs controls (saline, p53 DNA alone, and control NPs). Greater levels of apoptosis were also observed in p53NP-treated tumors. Overall, this led to significantly improved survival in p53NP-treated animals. NP-mediated p53 gene delivery slowed cancer progression and improved survival in an in vivo cancer model. One mechanism by which this was accomplished was disruption of tumor angiogenesis. We conclude that the NP-mediated sustained tumor p53 gene therapy can effectively be used for tumor growth inhibition.


Subject(s)
Genetic Therapy , Nanoparticles/therapeutic use , Neoplasms, Experimental , Neovascularization, Pathologic , Tumor Suppressor Protein p53 , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/therapeutic use
3.
Int J Pharm ; 270(1-2): 27-36, 2004 Feb 11.
Article in English | MEDLINE | ID: mdl-14726119

ABSTRACT

Rosin and Rosin-based polymers have diversified drug delivery applications achieving sustained/controlled release profiles. In this manuscript, two new Rosin derivatives were synthesized and evaluated for physicochemical properties, molecular weight, polydispersity and glass transition temperature. Plasticizer-free films prepared by solvent evaporation were tested for surface morphology, water vapour transmission and mechanical properties (tensile strength, percent elongation and modulus of elasticity). The films showed low tensile strength and high percent elongation values achieving smooth and uniform surface. The derivatives were further characterized for film coating by evaluating the release of a model drug (diclofenac sodium) from pellets coated with the rosin derivatives as retarding membrane. Drug release was sustained up to 10 h due to 10% (w/w) coat built up with the new rosin derivatives. Increase in coat-built-up further facilitated sustained release from coated forms. Film coating could be achieved without agglomeration of the pellets within a reasonable operating time. The present study proposes novel film forming materials with potential use in sustained drug delivery.


Subject(s)
Resins, Plant/chemistry , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Compounding , Hydrogen-Ion Concentration , Polymers/chemical synthesis , Polymers/chemistry , Resins, Plant/chemical synthesis , Solubility , Time Factors
4.
J Drug Target ; 10(6): 515-23, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12575742

ABSTRACT

Localized intramural delivery of sustained release biodegradable nanoparticles containing an antiproliferative agent could provide prolonged drug effect at the site of vascular injury that could inhibit the proliferation of smooth muscle cells and hence restenosis. The efficiency of arterial localization of nanoparticles is crucial in maximizing the drug effect in the target tissue. Therefore, the objective of the present study was to determine the comparative efficiency of the Dispatch and the Infiltrator cardiac infusion catheters to localize nanoparticles in the arterial wall. Following a standard balloon angioplasty procedure on the left anterior descending artery (LAD) in a porcine coronary model of restenosis, a suspension of nanoparticles containing a fluorescent marker was infused at the site of injury using either the Dispatch or the Infiltrator catheter. One hour following the infusion, animals were sacrificed and the nanoparticle levels in the LAD and other tissue were analyzed. The Dispatch catheter resulted in 3.3 folds greater efficiency of nanoparticle localization in the LAD than the Infiltrator catheter (309 +/- 124 vs. 93 +/- 43 microg/g of tissue, n = 6 for Dispatch and n = 5 for Infiltrator, p = 0.082, t-test). It is estimated that about 2% of the arterial volume can be displaced with the nanoparticle infusion. Fluorescence microscopy of the cross-sections of the LAD revealed greater fluorescence activity in the intimal layer with both the catheters, however the arteries infused using the Dispatch catheter demonstrated relatively higher degree of fluorescence activity in the medial and adventitial layers. The transmission electron microscopy of the arterial sections demonstrated infiltration of nanoparticles in the arterial wall and the histological analysis of the sections demonstrated no apparent damage to the endothelium due to the infusion of nanoparticles.


Subject(s)
Coronary Restenosis/drug therapy , Disease Models, Animal , Infusions, Intra-Arterial/methods , Nanotechnology/methods , Animals , Infusions, Intra-Arterial/instrumentation , Microspheres , Swine
5.
J Microencapsul ; 18(6): 723-32, 2001.
Article in English | MEDLINE | ID: mdl-11695637

ABSTRACT

The purpose of the study was to determine the synergistic adjuvant effect of sustained release biodegradable nanoparticles in combination with alum. Nanoparticles containing tetanus toxoid (TT) were formulated using a biodegradable polymer, polylactic polyglycolic acid co-polymer (50:50, molecular weight 100000). The immunization studies were carried out subcutaneously in rats. The results were expressed as mean serum anti-TT IgG levels. I'he nanoparticles demonstrated a TT loading of 4% w/w with mean particle diameter of 238 +/- 31 nm. The TT encapsulated in nanoparticles was released slowly under in vitro conditions, with 67.5% cumulative release occurring in 20 days. A single injection of TT-nanoparticles (TT dose = 10 Lf) mixed with TT-Alum (TT dose= 5 Lf) induced a four-fold greater mean serum anti-TT IgG response than a single injection of TT nanoparticles alone (TT dose=15Lf) (2235 +/- 310 vs. 539 +/- 49 microg/ml, mean +/- sem, p = >0.001). In addition, the mean immune response induced with the single injection of combination of nanoparticles and alum was comparable to the two injections of TT-alum alone (5 Lf each dose) given at 3 week intervals IgG = 1998 +/- 333 microg/ ml). Furthermore, the combination induced a peak immune response (IgG = 4215 +/- 546 microg/ml) as early as the first time point at 3 weeks post-immunization. In the case of a TT-alum alone, the animals showed a weaker immune response at 3 weeks and required a second dose of TT-alum to enhance the antibody response. The data thus suggest that the combination of TT-nanoparticles and TT-alum acts as a much better adjuvant than nanoparticles or alum alone. A rapid induction of immune response is useful to curb the spread of communicable diseases in the case of an outbreak.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Drug Compounding/methods , Tetanus Toxoid/administration & dosage , Animals , Biocompatible Materials , Biodegradation, Environmental , Capsules , Delayed-Action Preparations , Drug Carriers , Drug Synergism , Immunoglobulin G/blood , Injections, Subcutaneous , Lactic Acid , Nanotechnology , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Rats , Rats, Wistar , Tetanus Toxoid/immunology
6.
Crit Rev Eukaryot Gene Expr ; 11(1-3): 1-21, 2001.
Article in English | MEDLINE | ID: mdl-11693956

ABSTRACT

Angiogenesis is a novel approach for the therapy of various ischemia-related pathophysiologic conditions. Proangiogenic growth factors have shown promising results in preclinical studies using protein- and gene-based therapies. However, their success in clinical trials is hindered by the lack of an optimal delivery strategy that would provide sustained and localized levels of the growth factors in the diseased tissue. Targeted delivery of proangiogenic agents is expected to demonstrate therapeutic efficacy of growth factors at relatively lower doses, without the risk of systemic toxicity in terms of unwanted angiogenesis. To achieve the above objectives, various drug delivery systems are under investigation. This review describes the basic mechanism of action of growth factors, their current status in preclinical and clinical studies, and the issue of drug delivery.


Subject(s)
Arteriosclerosis/drug therapy , Collateral Circulation/drug effects , Growth Substances/therapeutic use , Angiopoietin-1 , Animals , Chick Embryo , Clinical Trials as Topic , Coronary Artery Disease/drug therapy , Drug Compounding , Drug Delivery Systems , Drug Evaluation, Preclinical , Endothelial Growth Factors/pharmacology , Endothelial Growth Factors/therapeutic use , Endothelium, Vascular/drug effects , Fibroblast Growth Factors/pharmacology , Forecasting , Genetic Therapy , Growth Substances/administration & dosage , Growth Substances/adverse effects , Hindlimb/blood supply , Humans , Hypotension/chemically induced , Ischemia/drug therapy , Lymphokines/pharmacology , Lymphokines/therapeutic use , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/therapeutic use , Mice , Neovascularization, Pathologic/chemically induced , Neovascularization, Physiologic/physiology , Rabbits , Rats , Receptors, Growth Factor/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
J Microencapsul ; 17(2): 215-25, 2000.
Article in English | MEDLINE | ID: mdl-10738697

ABSTRACT

In this study, the adjuvant effect of the sustained release biodegradable nanospheres (100-150 nm in diameter) has been compared with alum. Nanospheres were formulated using a biodegradable polylactic polyglycolic acid copolymer (PLGA, 50:50) containing Staphylococcal Enterotoxin B (SEB) toxoid as a model vaccine antigen. Systemic immune response of the nanospheres containing toxoid was studied in rabbits by subcutaneous immunization. The data demonstrated that approximately 30% of the toxoid activity was lost following its encapsulation into nanospheres. Under in vitro conditions, nanospheres demonstrated sustained release of the toxoid. However, only 20% of the antigenic toxoid was released over the first 2 weeks of the release study. Immunization of animals with equal doses of toxoid, either using nanospheres or alum induced a comparable systemic immune response (IgG, IgM and IgA). The immune response reached a maximum level at 7 weeks post-immunization, which then gradually declined with time. The booster dose of toxoid at 19 weeks, either using alum or nanospheres induced similar immune response in both the groups, but was greater than the primary immune response. The studies, thus, suggest that biodegradable nanospheres could be used as a vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/chemistry , Enterotoxins/chemistry , Enterotoxins/immunology , Staphylococcal Toxoid/chemistry , Staphylococcal Toxoid/immunology , Alum Compounds/administration & dosage , Animals , Biodegradation, Environmental , Dose-Response Relationship, Immunologic , Drug Carriers , Drug Stability , Electrophoresis, Polyacrylamide Gel , Enterotoxins/administration & dosage , Enzyme-Linked Immunosorbent Assay , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Injections, Subcutaneous , Male , Microscopy, Electron, Scanning , Microspheres , Particle Size , Rabbits , Staphylococcal Toxoid/administration & dosage
8.
J Pharm Sci ; 87(11): 1347-50, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9811488

ABSTRACT

In this paper we report a novel technique of DNA-polymer coating for gene transfer. A proprietary DNA polymer solution was used for thin-layer coating on a chromic gut suture as a model study. The coated sutures were characterized for physical properties such as coating thickness, mass of the DNA deposited on the suture, surface characteristics as determined by scanning electron microscopy, and in vitro DNA release characteristics under simulated physiologic conditions. The in vivo gene transfection using DNA-coated sutures was demonstrated in rat skeletal muscle and in canine atrial myocardium. A heat-stable human placental alkaline phosphatase (AP) plasmid was used as a marker gene. Incisions of 1 to 1.5 cm were made in the rat skeletal muscles or the canine atrial myocardium. The sites were closed with either the DNA-coated sutures or control sutures. Two weeks after the surgery, the tissue samples adjacent to the suture lines were retrieved and analyzed for AP activity. The DNA-coated sutures demonstrated a sustained release of the DNA under in vitro conditions, with an approximately 84% cumulative DNA release occurring in 26 days. An agarose gel electrophoresis of the DNA samples released from the suture demonstrated two bands, with the lower band corresponding to the input DNA (supercoiled). It seems that there was a partial transformation of the DNA from a supercoiled to an open circular form due to the polymer coating. The tissue sites, which received the DNA-coated sutures, demonstrated a significantly higher AP activity compared with the tissue sites that received control sutures. In the rat studies, the mean AP activity (square root of cpm/microgram protein) was 43.6 +/- 3.3 vs 20.6 +/- 2.1 (p = 0.001) at the control sites. Similarly, in the canine studies, the AP activity was 73.6 +/- 7.4 Vs 21.6 +/- 1.4 (p = 0.0009) at the control sites. Thus, our studies demonstrated a successful gene transfer using our DNA-polymer coating technique. This technique could be useful for coating sutures used in vascular and general surgery, and also for coating medical devices, such as stents, catheters, or orthopedic devices, to achieve a site-specific gene delivery.


Subject(s)
DNA/administration & dosage , Drug Delivery Systems , Genetic Therapy , Heart/physiology , Muscle, Skeletal/physiology , Animals , DNA/chemistry , Delayed-Action Preparations , Dogs , Drug Carriers , Humans , Male , Rats , Rats, Sprague-Dawley , Transfection
9.
J Pharm Sci ; 87(10): 1229-34, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9758682

ABSTRACT

Restenosis is the reobstruction of an artery following interventional procedures such as balloon angioplasty or stenting. Local pharmacotherapeutic approaches using controlled release systems are under investigation to inhibit the regional pathophysiologic process of restenosis. We have been investigating biodegradable nanoparticles (100 +/- 39 nm in diameter, mean +/- sd) for the local intra-arterial drug delivery. The purpose of this study was to investigate nanoparticle surface modifications (see Table 1) to enhance their arterial uptake. The PLGA (polylactic polyglycolic acid copolymer) nanoparticles were formulated by an oil-in-water emulsion solvent evaporation technique using a 2-aminochromone (U-86983, Upjohn and Pharmacia) (U-86) as a model antiproliferative agent. The various formulations of nanoparticles were evaluated for the arterial wall uptake by using an ex-vivo dog femoral artery model. The selected formulations were then tested in vivo in acute dog femoral artery and pig coronary artery models. The nanoparticles surface modified with a cationic compound, didodecyldimethylammonium bromide (DMAB), demonstrated 7-10-fold greater arterial U-86 levels compared to the unmodified nanoparticles in different ex-vivo and in-vivo studies. The mean U-86 levels were 10.7 +/- 1.7 microg/10 mg (dog) and 6.6 +/- 0.6 microg/10 mg (pig) in the artery segments ( approximately 2 cm) which were infused with the nanoparticles. The pig coronary studies further demonstrated that the infusion of nanoparticles with higher U-86 loading reduced the arterial U-86 levels, whereas increasing the nanoparticle concentration in the infusion solutions increased the arterial U-86 levels. The biodistribution studies in pigs following coronary arterial administration of nanoparticles demonstrated disposition of U-86 in the myocardium and distally in the liver and the lung. The mechanism of enhanced arterial uptake of the DMAB surface modified nanoparticles seems to be due to the alteration in the nanoparticle surface charge. The unmodified nanoparticles had a zeta potential of -27.8 +/- 0.5 mV (mean +/- sem, n = 5), whereas the DMAB modified nanoparticles demonstrated a zeta potential of +22.1 +/- 3.2 mV (mean +/- sem, n = 5). The adsorption of DMAB to the nanoparticle surface followed the Freundlich isotherm with binding capacity k = 28.1 microg/mg and affinity constant p = 2. 33. In conclusion, surface modified nanoparticles have potential applications for intra-arterial drug delivery to localize therapeutic agents in the arterial wall to inhibit restenosis.


Subject(s)
Chromones/pharmacokinetics , Coronary Vessels/metabolism , Femoral Artery/metabolism , Morpholines/pharmacokinetics , Animals , Arterial Occlusive Diseases/prevention & control , Biodegradation, Environmental , Chromones/administration & dosage , Chromones/chemistry , Dogs , Infusions, Intra-Arterial , Lactic Acid , Microspheres , Morpholines/administration & dosage , Morpholines/chemistry , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Surface Properties , Swine
10.
J Control Release ; 54(2): 201-11, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9724907

ABSTRACT

Biodegradable nanoparticles (NP) with a spherical diameter ranging from 70 to 160 nm were investigated for potential usefulness for the local intraluminal therapy of restenosis, the disease process responsible for arterial reobstruction following angioplasty. NPs containing a water-insoluble anti-proliferative agent U-86983 (U-86, Pharmacia and Upjohn, Kalamazoo, MI) were formulated from oil-water emulsions using biodegradable polymers such as poly(lactic acid-co-glycolic acid) (PLGA), and specific additives after particle formation, to enhance arterial retention using either heparin, didodecylmethylammonium bromide (DMAB), or fibrinogen, or combinations. Femoral and carotid arteries of male mongrel dogs were isolated in situ, and were then subjected to a balloon angioplasty. A NP suspension of a predetermined concentration was then infused into the artery for various durations. This was followed by a 30 min restoration of blood flow through the vessel. The arterial segments were excised and analyzed for drug levels. From the drug loading the NP and the drug levels in the artery, the quantity of nanoparticles retained was calculated and expressed as microgram per 10 mg dry arteries. In general, repeated short infusions of nanoparticle suspension (15 s x 4) were two-fold more effective in terms of higher arterial U-86 levels than a single prolonged infusion (60 s). A single 15 s infusion was not significantly different than a 60 s compared to non-modified NPs (39.2 +/- 2.5 and 49.1 +/- 2.4 vs. 21.5 +/- 0.6 micrograms/10 mg mean +/- s.e., respectively). A comparably enhanced NP uptake was noted with a combined heparin/DMAB modification. Increasing the concentration of NP in infusate from 5 to 30 mg ml-1 significantly increased arterial NP uptake level (from 22.5 +/- 3.5 to 83.7 +/- 1.4 micrograms/10 mg). Thus, the results support the view that modified nanoparticles along with optimized infusion conditions could enhance arterial wall drug concentrations of agents to treat restenosis.


Subject(s)
Arteries/drug effects , Chromones/therapeutic use , Drug Delivery Systems , Morpholines/therapeutic use , Angioplasty, Balloon , Animals , Arteries/chemistry , Arteries/metabolism , Biocompatible Materials/chemistry , Biodegradation, Environmental , Chromones/administration & dosage , Chromones/analysis , Coronary Disease/prevention & control , Disease Models, Animal , Dogs , Endothelium, Vascular/drug effects , Fibrinogen/analysis , Fibrinogen/pharmacology , Fibrinolytic Agents/analysis , Fibrinolytic Agents/pharmacology , Heparin/analysis , Heparin/pharmacology , Infusions, Intra-Arterial , Lactic Acid/chemistry , Male , Morpholines/administration & dosage , Morpholines/analysis , Particle Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry , Postoperative Complications/prevention & control
11.
J Cardiovasc Pharmacol ; 31(3): 449-55, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9514191

ABSTRACT

Atrial arrhythmias (atrial fibrillation or atrial flutter) after coronary artery bypass graft surgery are difficult to prevent or treat and often result in significant morbidity. Prior experimental studies by our group showed improved therapeutic efficacy for antiarrhythmic drugs delivered via controlled-release polymeric matrices implanted on the epicardial surface. These experiments were conducted to test the hypothesis that direct atrial epicardial administration of ibutilide from a controlled-release system (compared with intravenous administration) can reduce the inducibility of atrial flutter in the acute postoperative atrial myocardium. Polymeric sustained-release preparations were formulated by solvent casting of an ibutilide and polyurethane (Pellathane) solution in tetrahydrofurane. Multilayer solvent-casted coatings on pacing electrode wires were carried out to fabricate a sustained-release electrode system. In animal model studies, each dog underwent a thoracotomy, followed by a right atriotomy that was repaired. Induction of atrial flutter was attempted by burst pacing with the bipolar pacing catheter. Sinus rhythm was restored with overdrive pacing. After determining the induction rate (percentage) of atrial flutter in the baseline state, a stainless-steel wire coated with the drug-delivery system, 10% ibutilide/90% polyurethane (n = 7), or without drug (polyurethane coating without ibutilide, n = 5; control) was sewn onto the right atrium. Systemic intravenous administration of ibutilide (1.2 microg/kg/h) also was carried out in a separate group of animals after atriotomy (n = 5). For ibutilide (at an estimated dose of 1.2 microg/kg/h), the atrial-flutter results were 41.85 +/- 2.21% induction for baseline compared with 12.42 +/- 5.26% (p = 0.02) after the ibutilide wire implant. In the control dogs, atrial flutter was induced 29.4 +/- 4.7% at baseline and 25.2 +/- 5.1% after implantation of the control wire (p = 0.4). Ibutilide coronary venous serum concentrations at the end of the ibutilide-polyurethane electrode experiments were 2.25 +/- 0.2 ng/ml (mean +/- SEM) versus systemic levels that were below the limits of detection. Systemic intravenous ibutilide infusions had no effect on the inducibility of atrial flutter. In conclusion, an epicardial implantable electrode coating with an ibutilide controlled drug-release system significantly reduced the inducibility of atrial flutter in an experimental atriotomy model. These results suggest that atrial arrhythmias occurring after coronary bypass surgery may be prevented by local atrial administration of ibutilide from a controlled-release pacing electrode.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Atrial Fibrillation/prevention & control , Electrodes , Pacemaker, Artificial , Sulfonamides/administration & dosage , Animals , Anti-Arrhythmia Agents/therapeutic use , Dogs , Drug Delivery Systems , Microscopy, Electron, Scanning , Polymers , Sulfonamides/therapeutic use
12.
Am Heart J ; 133(3): 329-34, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9060802

ABSTRACT

Smooth-muscle cell proliferation in response to arterial injury represents an important etiologic factor in restenosis after angioplasty. Tyrphostin-47, a protein tyrosine kinase inhibitor, inhibits smooth-muscle cell proliferation in vitro. In this study tyrphostin-47 was incorporated into matrixes to determine whether prolonged local delivery would result in a reduction of neointimal proliferation after arterial injury in a rat carotid balloon-injury model. A polymer matrix (polylactic polyglycolic acid copolymer and pluronic gel F-127, mean matrix weight 7.83 +/- 0.39 mg) was loaded with tyrphostin-47 (25% w/w). Release studies demonstrated delivery of 11% of the incorporated drug over a 21-day release period. In cell culture, tyrphostin-47 released from the polymer matrix produced a reduction in smooth-muscle cell proliferation (p < 0.0007). Balloon denudation injury of the left common carotid artery of 34 animals was performed. In 12 animals, polymer matrixes containing tyrphostin-47 were wrapped around the injured arteries to provide prolonged drug delivery (estimated dosage 28 micrograms/kg/24 hr); in 10 animals a polymer matrix without tyrphostin-47 was implanted; and in 12 animals only balloon injury was performed. The mean neointimal cross-sectional areas, luminal areas, and intima/media ratios were not significantly different among animals receiving local treatment with tyrphostin-47, sham polymer after injury, or balloon injury without polymer implantation. We conclude that despite inhibition of smooth-muscle cell proliferation by tyrphostin-47 in vitro, sustained local delivery of this tyrosine kinase inhibitor does not result in a reduction of neointimal proliferation in the rat carotid injury model.


Subject(s)
Carotid Artery, Common/pathology , Drug Delivery Systems , Muscle, Smooth/cytology , Nitriles/administration & dosage , Phenols/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrphostins , Animals , Cell Division/drug effects , Cells, Cultured , Disease Models, Animal , Male , Polymers , Rats , Rats, Sprague-Dawley , Tunica Intima/pathology , Tunica Intima/physiology
13.
Pharm Res ; 14(11): 1568-73, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9434276

ABSTRACT

PURPOSE: To study the uptake of biodegradable microparticles in Caco-2 cells. METHODS: Biodegradable microparticles of polylactic polyglycolic acid co-polymer (PLGA 50:50) of mean diameters 0.1 micron, 1 micron, and 10 microns containing bovine serum albumin as a model protein and 6-coumarin as a fluorescent marker were formulated by a multiple emulsion technique. The Caco-2 cell monolayers were incubated with each diameter microparticles (100 micrograms/ml) for two hours. The microparticle uptake in Caco-2 cells was studied by confocal microscopy and also by quantitating the 6-coumarin content of the microparticles taken up by the cells. The effects of microparticle concentration, and incubation time and temperature on microparticle cell uptake were also studied. RESULTS: The study demonstrated that the Caco-2 cell microparticle uptake significantly depends upon the microparticle diameter. The 0.1 micron diameter microparticles had 2.5 fold greater uptake on the weight basis than the 1 micron and 6 fold greater than the 10 microns diameter microparticles. Similarly in terms of number the uptake of 0.1 micron diameter microparticles was 2.7 x 10(3) fold greater than the 1 micron and 6.7 x 10(6) greater than the 10 microns diameter microparticles. The efficiency of uptake of 0.1 micron diameter microparticles at 100 micrograms/ml concentration was 41% compared to 15% and 6% for the 1 micron and the 10 microns diameter microparticles, respectively. The Caco-2 cell microparticle (0.1 micron) uptake increased with concentration in the range of 100 micrograms/ml to 500 micrograms/ml which then reached a plateau at higher concentration. The uptake of microparticles increased with incubation time, reaching a steady state at two hours. The uptake was greater at an incubation temperature of 37 degrees C compared to at 4 degrees C. CONCLUSIONS: The Caco-2 cell microparticle uptake was microparticle diameter, concentration, and incubation time and temperature dependent. The small diameter microparticles (0.1 micron) had significantly greater uptake compared to larger diameter microparticles. The results thus suggest that the mechanism of uptake of microparticles in Caco-2 cell is particle diameter dependent. Caco-2 cells are used as an in vitro model for gastrointestinal uptake, and therefore the results obtained in these studies could be of significant importance in optimizing the microparticle-based oral drug delivery systems.


Subject(s)
Caco-2 Cells/metabolism , Cell Membrane Permeability , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polymers/chemistry , Polymers/pharmacokinetics , Animals , Cattle , Humans , Intestinal Absorption , Microscopy, Confocal , Microspheres , Models, Biological , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Serum Albumin, Bovine/metabolism
14.
Pharm Res ; 13(12): 1838-45, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8987081

ABSTRACT

PURPOSE: To investigate the effect of microparticle size on gastrointestinal tissue uptake. METHODS: Biodegradable microparticles of various sizes using polylactic polyglycolic acid (50:50) co-polymer (100 nm, 500 nm, 1 micron, and 10 microns) and bovine serum albumin as a model protein were formulated by water-in-oil-in-water emulsion solvent evaporation technique. The uptake of microparticles was studied in rat in situ intestinal loop model and quantitatively analyzed for efficiency of uptake. RESULTS: In general, the efficiency of uptake of 100 nm size particles by the intestinal tissue was 15-250 fold higher compared to larger size microparticles. The efficiency of uptake was dependent on the type of tissue, such as Peyer's patch and non patch as well as on the location of the tissue collected i.e. duodenum or ileum. Depending on the size of microparticles, the Peyer's patch tissue had 2-200 fold higher uptake of particles than the non-patch tissue collected from the same region of the intestine. Histological evaluation of the tissue sections demonstrated that 100 nm particles were diffused throughout the submucosal layers while the larger size nano/microparticles were predominantly localized in the epithelial lining of the tissue. CONCLUSIONS: There is a microparticle size dependent exclusion phenomena in the gastrointestinal mucosal tissue with 100 nm size particles showing significantly greater tissue uptake. This has important implications in designing of nanoparticle-based oral drug delivery systems, such as an oral vaccine system.


Subject(s)
Biocompatible Materials/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Lactic Acid , Particle Size , Polyglycolic Acid , Polymers/pharmacology , Animals , Biodegradation, Environmental , Duodenum/anatomy & histology , Duodenum/metabolism , Ileum/anatomy & histology , Ileum/metabolism , In Vitro Techniques , Male , Peyer's Patches/anatomy & histology , Peyer's Patches/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
15.
Circulation ; 94(6): 1441-8, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8823004

ABSTRACT

BACKGROUND: Several perfusion balloon catheters are under investigation for local drug delivery; however, sustained tissue drug levels are difficult to achieve with these techniques. To overcome this problem, sustained-release, biodegradable nanoparticles represent a potential alternative for prolonged local delivery. METHODS AND RESULTS: A biodegradable polylactic-polyglycolic acid (PLGA) copolymer was used to formulate nanoparticles. Fluorescent-labeled nanoparticles were intraluminally administered in a single, 180-second infusion after balloon injury in the rat carotid model. Localization and retention at different time points and biocompatibility of nanoparticles were evaluated. To evaluate the potential of the system in the prevention of neointimal formation, dexamethasone was incorporated into the particles and delivered locally as above. Nanoparticles were seen in the three layers of the artery at 3 hours and 24 hours. At 3 days, they were mainly present in the adventitial layer, decreasing at 7 days, with no fluorescent activity at 14 days. The PLGA nanoparticles appeared to be fully biocompatible. In the dexamethasone nanoparticle study, a significant amount of dexamethasone was present in the treated segment for up to 14 days after a single infusion, with no plasma levels detected after the first 3 hours. There was a 31% reduction in intima-media ratio in animals treated with local dexamethasone nanoparticles compared with control. CONCLUSIONS: Nanoparticles successfully penetrated into the vessel wall and persisted for up to 14 days after a short, single intraluminal infusion. Local administration of nanoparticles with incorporated dexamethasone significantly decreased neointimal formation. This methodology appears to have important potential for clinical applications in local drug delivery.


Subject(s)
Angioplasty, Balloon, Coronary , Drug Delivery Systems , Microspheres , Postoperative Care , Animals , Biocompatible Materials , Biodegradation, Environmental , Carotid Arteries/drug effects , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Dexamethasone/pharmacology , Injections, Intra-Arterial , Injections, Intraperitoneal , Male , Neovascularization, Pathologic/prevention & control , Polymers , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tunica Intima/drug effects
16.
Drug Deliv ; 3(3): 137-42, 1996.
Article in English | MEDLINE | ID: mdl-26790908

ABSTRACT

Cardiac implants using drug-polymer systems for the release of anti-arrhythmic agents directly to the myocardium have been successfully utilized in experimental studies for preventing and treating arrhythmias. This approach is hypothesized to be optimal since anti-arrhythmic agents are provided directly to the heart, and therefore the possibility of systemic side effects is reduced. Furthermore, anti-arrhythmic agents with poor oral bioavailability or with first-pass clearance characteristics following intravenous administration, such as the class III agent ibutilide, are optimally used by the direct cardiac route of administration. Cardiac controlled release implants have been demonstrated to be effective for optimizing the therapy of ventricular tachycardia, ventricular fibrillation, and atrial flutter. Monolithic matrices have been used for fixed rate release kinetics, and modulation of release has been possible through the use of iontophoretic drug delivery systems. Future implants will interface with forefront strategies in tissue engineering and molecular genetics to provide optimal therapy of the diseased arrhythmogenic myocardium.

17.
Clin Pharmacokinet ; 29(1): 1-5, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7586894

ABSTRACT

Conventional antiarrhythmia therapy by oral or intravenous routes of administration is often ineffective and results in drug-associated complications and toxicity. In addition, poor bioavailability and a high first-pass effect limit therapeutic applications of several investigational antiarrhythmic compounds, which are otherwise more potent and less toxic than available agents. The regional nature of the several cardiac diseases, such as ischaemia, restenosis or heart valve calcification, may require a high concentration of drug at the location of the disease, which by conventional routes may not be attainable due to systemic toxicity of the drug. Localised cardiac delivery of antiarrhythmic agents, based on drug-polymer implants, may have several advantages, including enhanced drug effects and reduced systemic drug toxicity. Computer-assisted automated feedback systems may further enhance the usefulness of this therapy in the clinical setting. Before clinical application of this method of drug delivery further study will be required, but it is hypothesised that pharmacokinetic variability for drugs delivered in this manner will be reduced and therefore efficacy and toxicity will be more predictable.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/pharmacokinetics , Drug Delivery Systems , Heart/drug effects , Myocardium/metabolism , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Biological Availability , Delayed-Action Preparations/standards , Drug Implants , Feedback , Iontophoresis
19.
Circ Res ; 76(4): 505-13, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7895327

ABSTRACT

The process of restenosis after arterial balloon dilatation has been demonstrated to involve smooth muscle cell hyperplasia. Initial reports with antisense oligonucleotides directed against the proto-oncogene c-myb suggest marked in vitro specificity and in vivo efficacy. In the present study, we sought to confirm and extend the hypothesis that antisense to c-myb results in a specific antiproliferative effect with a comprehensive assessment by using different oligonucleotide preparations, different species, and tissue and cellular uptake experiments. Phosphorothioate-protected oligonucleotides representing the appropriate sequence for antisense to c-myb and multiple controls were used to inhibit proliferation of platelet-derived growth factor- and fetal bovine serum-stimulated rat, dog, and human aortic smooth muscle cells in vitro and neointimal proliferation in the rat carotid injury model. In vitro experiments using identical culture conditions in rat, dog, and human aortic smooth muscle cells failed to show specificity as well as consistency in growth inhibitory effects that could be attributed to an antisense mechanism. Proliferation of smooth muscle cell growth in culture was consistently inhibited with oligomers containing a contiguous 4-guanosine residue motif. In vivo, the rat carotid injury neointimal hyperplasia was similar for antisense c-myb (0.095 +/- 0.009 mm2) and sense c-myb (0.090 +/- 0.009 mm2). Fluorescent-labeled oligonucleotides were present in tissue after local delivery via pluronic gel, and their activity rapidly declined over a 72-hour period. Our findings point to the potential nonspecificity and lack of consistency of the antisense oligonucleotide to c-myb in vitro and in vivo.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Arteries/injuries , Cell Division/drug effects , Muscle, Smooth, Vascular/cytology , Oligonucleotides, Antisense/pharmacology , Oncogenes , Angioplasty, Balloon/adverse effects , Animals , Aorta , Carotid Arteries/pathology , Carotid Artery Injuries , Carotid Stenosis/therapy , Dogs , Histological Techniques , Humans , Hyperplasia , In Vitro Techniques , Mice , Oligonucleotides, Antisense/genetics , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley , Recurrence , Sensitivity and Specificity
20.
Proc Natl Acad Sci U S A ; 92(7): 2612-6, 1995 Mar 28.
Article in English | MEDLINE | ID: mdl-7708693

ABSTRACT

Cardiac arrhythmias are a frequent cause of death and morbidity. Conventional antiarrhythmia therapy involving oral or intravenous medication is often ineffective and complicated by drug-associated side effects. Previous studies from our laboratory have demonstrated the advantages of cardiac drug-polymer implants for enhanced efficacy for cardiac arrhythmia therapy compared with conventional administration. However, these studies were based on systems that deliver drugs at a fixed release rate. Modulation of the drug delivery rate has the advantage of regulating the amount of the drug delivered depending upon the disease state of the patient. We hypothesized that iontophoresis could be used to modulate cardiac drug delivery. In this study, we report our investigations of a cardiac drug implant in dogs that is capable of iontophoretic modulation of the administration of the antiarrhythmic agent sotalol. We used a heterogeneous cation-exchange membrane (HCM) as an electrically sensitive and highly efficient rate-limiting barrier on the cardiac-contacting surface of the implant. Thus, electric current is passed only through the HCM and not the myocardium. The iontophoretic cardiac implant demonstrated in vitro drug release rates that were responsive to current modulation. In vivo results in dogs have confirmed that iontophoresis resulted in regional coronary enhancement of sotalol levels with current-responsive increases in drug concentrations. We also observed acute current-dependent changes in ventricular effective refractory periods reflecting sotalol-induced refractoriness due to regional drug administration. In 30-day dog experiments, iontophoretic cardiac implants demonstrated robust sustained function and reproducible modulation of drug delivery kinetics.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Dogs , Iontophoresis/methods , Sotalol/administration & dosage , Animals , Coronary Circulation , Drug Carriers , Drug Implants , Iontophoresis/instrumentation , Sotalol/blood , Sotalol/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...