Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Appl Biomater Biomech ; 8(1): 42-51, 2010.
Article in English | MEDLINE | ID: mdl-20740421

ABSTRACT

PURPOSE: The aim of the present study was to synthetize and characterize novel sol-gel organic-inorganic hybrid materials to be used for controlled drug delivery application. MATERIALS AND METHODS: Organic-inorganic hybrid class I materials based on poly(epsilon-caprolactone) (PCL 6, 12, 24 and 50 wt%) and zirconia-yttria (ZrO2-5%Y2O3) were synthesized by a sol-gel method, from a multicomponent solution containing zirconium propoxide [Zr(OC2H7)4], yttrium chloride (YCl3), PCL, water and chloroform (CHCl3). The structure of the hybrids was obtained by means of hydrogen bonds between the Zr-OH group (H-donor) in the sol-gel intermediate species and the carboxylic group (H-acceptor) in the repeating units of the polymer. RESULTS: The presence of hydrogen bonds between organic-inorganic components of the hybrid materials was suggested by Fourier transform infrared (FTIR) analysis, and strongly supported by solid-state NMR. A single-step, sol-gel process was then used to precipitate microspheres containing ketoprofen or indomethacin for controlled drug delivery applications. Release kinetics in a simulated body fluid (SBF) were subsequently investigated. The amount of drug released was detected by UV-VIS spectroscopy. Pure anti-inflammatory agents exhibited linear release with time, in contrast drugs entrapped in the organic-inorganic hybrids were released with a logarithmic time dependence, starting with an initial burst effect followed by a gradual decrease. CONCLUSIONS: The synthesis of amorphous materials containing drugs, obtained by sol-gel methods, helps to devise new strategies for controlled drug delivery system design.


Subject(s)
Infusion Pumps, Implantable , Polyesters/chemistry , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Indomethacin/administration & dosage , Indomethacin/therapeutic use , Inorganic Chemicals/chemistry , Ketoprofen/administration & dosage , Ketoprofen/therapeutic use , Kinetics , Magnetic Resonance Spectroscopy/methods , Organic Chemicals/chemistry , Oxides/chemistry , Phase Transition , Spectroscopy, Fourier Transform Infrared/methods , Zirconium/chemistry
2.
J Cell Physiol ; 221(2): 440-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19650107

ABSTRACT

The methyl ester prodrug of N(omega)-nitro-L-arginine (L-NAME) has been reported to exert anticancer effects against several human tumors, including thyroid carcinoma, by inhibiting nitric oxide synthase (NOS). However, chronic administration of L-NAME has often led to adverse events causing cardiovascular alterations due to its potential toxic effect. Here we report for the first time the synthesis of the galactosyl ester prodrug of N(omega)-nitro-L-arginine, NAGAL, a prodrug capable of inhibiting NOS more efficiently and with fewer adverse events than its parent drug. For this purpose RO82-W-1, a thyroid cell line derived from human follicular carcinoma, was used. MTT test results showed that NAGAL affected cell viability to a significantly greater extent than did L-NAME. Moreover, fluorescence activated cell sorter (FACS) analyses revealed that NAGAL, compared to L-NAME, was able to reduce nitric oxide (NO) production as well as increase the percentage of apoptotic thyreocytes. Western blot further confirmed the reduction in NOS-II expression by NAGAL. Finally, by using the LC-MS technique, we found that NAGAL elicited a higher increase in N(omega)-nitro-L-arginine (NA) concentration than did L-NAME. Thus, this study suggests that NAGAL could be considered a potential therapeutic tool for those pathologies involving an overproduction of NO, including thyroid carcinoma.


Subject(s)
Adenocarcinoma, Follicular/pathology , Galactose/pharmacology , Nitroarginine/pharmacology , Thyroid Neoplasms/pathology , Apoptosis/drug effects , Biological Assay , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Galactose/chemistry , Humans , Nitric Oxide/metabolism , Nitroarginine/chemical synthesis , Nitroarginine/chemistry , Nitroarginine/metabolism , Time Factors
3.
J Pharmacol Exp Ther ; 331(1): 212-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19602550

ABSTRACT

With the help of single-cell microflorimetry, (45)Ca(2+) radiotracer fluxes, and patch-clamp in whole-cell configuration, we examined the effect of the amiloride derivative 3-amino-6-chloro-5-[(4-chloro-benzyl)amino]-N-[[(2,4-dimethylbenzyl)amino]iminomethyl]-pyrazinecarboxamide (CB-DMB) on the activity of the three isoforms of the Na(+)/Ca(2+) exchanger (NCX) and on several other membrane currents including voltage- and pH-sensitive ones. This amiloride analog suppressed the bidirectional activity of all NCX isoforms in a concentration-dependent manner. The IC(50) values of CB-DMB were in the nanomolar range for the outward and the inward components of the bidirectional NCX1, NCX2, and NCX3 activity. Deletion mutagenesis showed that CB-DMB inhibited NCX activity mainly at level of the f-loop but not through the interaction with Gly833 located at the level of the alpha(2) repeat. On the other hand, CB-DMB suppressed in the micromolar range the other plasma membrane currents encoded by voltage-dependent Ca(2+) channels, tetrodotoxin-sensitive Na(+) channels, and pH-sensitive ASIC1a. Collectively, the data of the present study showed that CB-DMB, when used in the nanomolar range, is one of the most potent compounds that can block the activity of the three NCX isoforms when they work both in the forward and in the reverse modes of operation without interfering with other ionic channels.


Subject(s)
Amiloride/analogs & derivatives , Sodium-Calcium Exchanger/antagonists & inhibitors , Amiloride/chemistry , Amiloride/pharmacology , Animals , Cell Line , Cells, Cultured , Cricetinae , Dogs , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/physiology , Rats , Sodium-Calcium Exchanger/physiology , Transfection/methods
4.
Neuropharmacology ; 56(3): 637-46, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19071141

ABSTRACT

It is presently unclear whether the antiseizure effects exerted by NSAIDs are totally dependent on COX inhibition or not. To clarify this point we investigated whether 7-methyl-2-phenylimidazo[1,2-b]pyridazine-3-carboxylic acid (DM1) and 6-methoxy-2-phenylimidazo[1,2-b]pyridazine-3-carboxylic acid (DM2), two imidazo[1,2-b]pyridazines structurally related to indomethacin (IDM) but ineffective in blocking COXs, retain IDM antiabsence activity. When administered by intraperitoneal injection in WAG/Rij rats, a rat strain which spontaneously develops SWDs, both DM1 and DM2 dose-dependently suppressed the occurrence of these seizures. Importantly, these compounds were both more potent in suppressing SWD occurrence than IDM. As T-type channel blockade is considered a mechanism of action common to many antiabsence drugs we explored by whole cell patch clamp electrophysiology in stably transfected HEK-293 the effect of DM1 and DM2 on Ca(V)3.1 channels, the T-type channel subtype preferentially expressed in ventrobasal thalamic nuclei. Both these compounds dose-dependently suppressed the currents elicited by membrane depolarization in these cells. A similar T-type blocking effect was also observed when the cells were exposed to IDM. In conclusion, DM1 and DM2 whilst inactive on COXs, are potent antiabsence drugs. This suggests that compounds with structural features typical of NSAIDs may exert antiepileptic activity independently from COX inhibition and possibly by a direct interaction with T-type voltage-dependent Ca(2+) channels.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticonvulsants/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/drug effects , Epilepsy, Absence/drug therapy , Imidazoles/therapeutic use , Indomethacin/analogs & derivatives , Indomethacin/therapeutic use , Pyridazines/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Disease Models, Animal , Electroencephalography , Imidazoles/pharmacology , In Vitro Techniques , Indomethacin/pharmacology , Male , Prostaglandin-Endoperoxide Synthases/metabolism , Pyridazines/pharmacology , Random Allocation , Rats , Rats, Inbred Strains
5.
J Med Chem ; 49(16): 4826-33, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16884294

ABSTRACT

Nitric oxide (NO) is critical for the normal physiological regulation of the nervous system and other tissues. L-Arginine, but not D-arginine, is the natural substrate for nitric oxide synthase (NOS), for it is enzymatically converted to NO and L-citrulline. However, recent evidence suggests that D-arginine can also produce NO and NO-derivatives via a different pathway. The aim of the present paper was to raise NO levels in the cells by increasing the cell permeation of its precursors. To this aim, two galactosyl prodrugs, L-arginine-D-galactos-6'-yl ester (L-ArgGal) and D-arginine-D-galactos-6'-yl ester (D-ArgGal) were synthesized. Remarkably, using the HPLC-ESI/MS technique, we found that L-ArgGal and D-ArgGal prodrugs both increased the concentration levels of L- and D-arginine and their derivatives in pituitary GH3 cells. Furthermore, we found that D-ArgGal (1) penetrated cell membranes more rapidly than its precursor D-arginine, (2) released arginine more slowly and in greater amounts than L-ArgGal, and (3) produced much higher levels of DAF-2 monitored NO and nitrite than did L-ArgGal under the same experimental conditions. In conclusion, these results indicate that an increase in the cell permeation of L- and D-arginine by L-ArgGal and D-ArgGal can lead to an increase in NO levels.


Subject(s)
Arginine/analogs & derivatives , Arginine/chemical synthesis , Galactose/analogs & derivatives , Galactose/chemical synthesis , Nitric Oxide/biosynthesis , Prodrugs/chemical synthesis , Animals , Arginine/pharmacology , Cattle , Cell Line, Tumor , Cell Membrane Permeability , Cells, Cultured , Chromatography, High Pressure Liquid , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Galactose/pharmacology , Nitrites/metabolism , Pituitary Gland/cytology , Prodrugs/pharmacology , Rats , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Structure-Activity Relationship
6.
J Drug Target ; 11(1): 25-36, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12852438

ABSTRACT

Novel glycosyl derivatives of dopamine and L-dopa (I-IV) are synthesized in order to overcome the problem of blood-brain barrier low permeability of dopamine and of low bioavailability of its precursor L-dopa. Esters synthesized link dopamine and L-dopa, by a succinyl linker, to C-3 position of glucose (I and II) and to C-6 of galactose (II and IV). The chemical and enzymatic stabilities of esters synthesized were evaluated in order to determine both their stability in aqueous medium and their feasibility in undergoing enzymatic cleavage by rat plasma to regenerate the original drug. Furthermore, we have shown the central effects of esters I-IV on classic dopaminergic models, such as morphine induced locomotion and reserpine-induced hypolocomotion. From the result obtained compounds I-IV appeared moderately stable in a pH 7.4 buffered solution and in rat plasma. Furthermore, pharmacological studies showed that both dopamine derivatives (I and II) were equiactive in reversing reserpine-induced hypolocomotion in rats, and both were more active than L-dopa or ester III and IV, while II and III were more potent in reducing morphine-induced locomotion than I and IV. The minimal vascular effects of these derivatives allow us to underline the possibility to use them in pathologies, such as Parkinson disease, characterised by an evident decreasing of dopamine concentration in the brain.


Subject(s)
Antiparkinson Agents/pharmacology , Dopamine/pharmacology , Levodopa/pharmacology , Motor Activity/drug effects , Prodrugs/pharmacology , Animals , Antiparkinson Agents/chemical synthesis , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Dopamine/analogs & derivatives , Dopamine/chemical synthesis , Drug Stability , Galactose/analogs & derivatives , Galactose/chemical synthesis , Galactose/pharmacology , Glucose/analogs & derivatives , Glucose/chemical synthesis , Glucose/pharmacology , Levodopa/analogs & derivatives , Levodopa/chemical synthesis , Male , Mice , Motor Activity/physiology , Prodrugs/chemical synthesis , Rats , Rats, Wistar
7.
Eur J Pharm Sci ; 62: 33-9, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24854456

ABSTRACT

N-Palmitoylethanolamide (PEA) is emerging as a novel therapeutic agent in the treatment of neuropathic pain and neurodegenerative diseases. Unfortunately, PEA poorly reaches the central nervous system (CNS), after peripheral administration, since it is inactivated through intracellular hydrolysis by lipid amidases. Since prodrug approach is one of the most popular methods used to increase cell permeability, the aim of this paper consists in the synthesis of a new galactosyl prodrug of PEA, the palmitoylethanolamide-succinamyl-D-galactos-6'-yl ester (PEAGAL). Biological experiments both in neuroblastoma and in C6 glioma cells, together with quantitative analyses performed through a LC-MS-MS technique, demonstrate the better efficacy of PEAGAL compared to PEA and its higher cell permeation. Our results encourage further experiments in animal models of neuropathic pain and of neurological disorders and/or neurodegenerative diseases, in order to promote a more effective peripherally administrated derivative of PEA.


Subject(s)
Analgesics/pharmacology , Galactose/analogs & derivatives , Neuroprotective Agents/pharmacology , Palmitates/pharmacology , Prodrugs/pharmacology , Amides , Analgesics/chemical synthesis , Analgesics/chemistry , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/drug effects , Drug Stability , Ethanolamines/metabolism , Galactose/chemical synthesis , Galactose/chemistry , Galactose/pharmacology , Humans , Hydrogen-Ion Concentration , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Nitrites/metabolism , Oxidopamine/toxicity , Palmitates/chemical synthesis , Palmitates/chemistry , Palmitic Acids/metabolism , Permeability/drug effects , Prodrugs/chemical synthesis , Prodrugs/chemistry
8.
Int J Nanomedicine ; 7: 5683-91, 2012.
Article in English | MEDLINE | ID: mdl-23180963

ABSTRACT

PURPOSE: Epidemiological, clinical, and laboratory studies have suggested that ibuprofen, a commonly used nonsteroidal anti-inflammatory drug, inhibits the promotion and proliferation of certain tumors. Recently, we demonstrated the antiproliferative effects of ibuprofen on the human gastric cancer cell line MKN-45. However, high doses of ibuprofen were required to elicit these antiproliferative effects in vitro. The present research compared the antiproliferative effects of ibuprofen delivered freely and released by poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) in MKN-45 cells. METHODS: MKN-45 human gastric adenocarcinoma cells were treated with ibuprofen-loaded PLGA NPs. The proliferation of MKN-45 cells was then assessed by cell counting. The uptake of NPs was imaged by fluorescence microscopy and flow cytometry. The release of ibuprofen from ibuprofen-loaded PLGA NPs in the cells was evaluated by gas chromatography-mass spectrometry. RESULTS: Dramatic inhibition of cellular proliferation was observed in cells treated with ibuprofen-loaded PLGA NPs versus those treated with free ibuprofen at the same concentration. The localization of NPs was cytoplasmic. The initiation of ibuprofen release was rapid, commencing within 2 hours, and then increased slowly over time, reaching a maximum concentration at 24 hours. The inhibition of proliferation was confirmed to be due to the intracellular release of ibuprofen from the NPs. Using PLGA NPs as carriers, ibuprofen exerted an antiproliferative activity at concentrations > 100 times less than free ibuprofen, suggesting greater efficiency and less cellular toxicity. In addition, when carried by PLGA NPs, ibuprofen more quickly induced the expression of transcripts involved in proliferation and invasiveness processes. CONCLUSION: Ibuprofen exerted an antiproliferative effect on MKN-45 cells at low concentrations. This effect was achieved using PLGA NPs as carriers of low doses of ibuprofen.


Subject(s)
Ibuprofen/administration & dosage , Lactic Acid/chemistry , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Polyglycolic Acid/chemistry , Stomach Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Ibuprofen/chemistry , Nanocapsules/ultrastructure , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Stomach Neoplasms/pathology , Treatment Outcome
9.
Curr Top Med Chem ; 11(18): 2288-98, 2011.
Article in English | MEDLINE | ID: mdl-21671867

ABSTRACT

D-galactose is a simple and natural compound that has mainly been exploited in prodrug strategies. Galactosyl prodrugs can be considered a good approach to reach different goals in clinical drug application, especially when traditional drugs are likely to fail therapeutically owing to reasons such as the lack of site specificity, toxicity, and chemical instability. Indeed, of paramount importance is their ability to increase the selectivity of the parent compound, a phenomenon that helps to reduce the incidence of adverse effects, while preserving intact the pharmacodynamic features of the parent drug. Study results have varied according to the type of linkage between the drug and the hydroxyl group exploited. By working with these parameters, researchers have been able not only to generate selective pharmacological targeting of brain, liver, and cancerous cells, but also to improve cellular permeability as well as the pharmacokinetic profile of parent drugs. This review describes the broad spectrum of possibilities for exploiting D-galactose as a vector for prodrug design and the synthetic strategies that allow its realization.


Subject(s)
Drug Carriers/metabolism , Drug Design , Galactose/chemistry , Galactose/metabolism , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Tissue Distribution
10.
Nanoscale ; 3(12): 5110-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22037807

ABSTRACT

The development of fluorescent biolabels for specific targeting and controlled drug release is of paramount importance in biological applications due to their potential in the generation of novel tools for simultaneous diagnosis and treatment of diseases. Dopamine is a neurotransmitter involved in several neurological diseases, such as Parkinson's disease and attention deficit hyperactivity disorder (ADHD), and the controlled delivery of its agonists already proved to have beneficial effects both in vitro and in vivo. Here, we report the synthesis and multiple functionalization of highly fluorescent CdSe/CdS quantum rods for specific biolabeling and controlled drug release. After being transferred into aqueous media, the nanocrystals were made highly biocompatible through PEG conjugation and covered by a carbohydrate shell, which allowed specific GLUT-1 recognition. Controlled attachment of dopamine through an ester bond also allowed hydrolysis by esterases, yielding a smart nanotool for specific biolabeling and controlled drug release.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Dopamine Agents/pharmacology , Dopamine/pharmacology , Drug Delivery Systems/methods , Parkinson Disease/drug therapy , Quantum Dots , Cadmium/chemistry , Cadmium Compounds/chemistry , Cell Line, Tumor , Dopamine/chemistry , Dopamine Agents/chemistry , Fluorescent Dyes/chemistry , Glucose Transporter Type 1/agonists , Glucose Transporter Type 1/metabolism , Humans , Polyethylene Glycols/chemistry , Selenium/chemistry , Staining and Labeling/methods , Sulfides/chemistry
11.
Eur J Pharmacol ; 656(1-3): 52-62, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21296071

ABSTRACT

This study has investigated whether the galactosyl ester prodrug of N(ω)-nitro-L-arginine (NAGAL), shows enhanced analgesic efficacy in healthy mice and in models of visceral and neuropathic pain: the writhing test and the spared nerve injury (SNI), respectively. NAGAL was compared to methyl ester pro-drug of N(ω)-nitro-l-arginine (L-NAME), a widely exploited non-specific nitric oxide synthase (NOS) inhibitor, for analgesic potential. The writhing test revealed that the ED(50) value, along with the 95% confidence limit (CL) was 3.82 (1.77-6.04) mg/kg for NAGAL and, 36.75 (20.07-68.37) mg/kg for L-NAME. Notably, NAGAL elicited a greater anti-allodynic effect than L-NAME did in neuropathic mice. Biomolecular and morphological studies revealed that spared nerve injury increased the expressions of pro-inflammatory enzymes (caspase-1) and two glial cell biomarkers: integrin alpha M (ITGAM) and glial fibrillary acidic protein (GFAP) in the spinal cord. Finally, GLUT-3, an isoform of the hexose transporters capable to bind NAGAL and inducible NOS (iNOS), were found to be over-expressed in the activated astrocytes of the spinal cord of neuropathic mice. NAGAL administration normalized expression levels of these biomarkers. NAGAL showed a greater efficacy in inhibiting visceral pain and allodynia than L-NAME possibly by a greater cell permeation through the hexose transporter which is highly over-expressed by activated glia.


Subject(s)
Galactose/metabolism , Hyperalgesia/drug therapy , Neuralgia/pathology , Neuroglia/drug effects , Nitroarginine/metabolism , Nitroarginine/pharmacology , Prodrugs/metabolism , Analgesics/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/pathology , Blood Pressure/drug effects , Caspases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Glucose Transporter Type 3/metabolism , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Neuralgia/metabolism , Neuralgia/physiopathology , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/pathology , Nitric Oxide Synthase Type II/metabolism , Nitroarginine/therapeutic use , Psychomotor Performance/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Time Factors
12.
Neuropharmacology ; 58(3): 660-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19944111

ABSTRACT

The 6-methoxy-2-phenylimidazo[1,2-b]pyridazine-3-carboxylic acid, DM2, exerts anti-absence activity and blocks Cav3.1 channel, a T-type voltage-dependent Ca(2+) channel subtype, in vitro. The current study investigated the effect of intra-ventrolateral periaqueductal grey (VLPAG) administration of DM2 on formalin-induced nocifensive responses in rats. In addition, the effect of intra-VLPAG microinjection of DM2 on the ongoing and tail flick-related activities of rostral ventromedial medulla (RVM) cell population was also investigated. Formalin was injected subcutaneously into the dorsal surface of the hind paws of awake rats. We found that DM2 reduced nocifensive responses in the late phase of the formalin test. Moreover, in the RVM, the intra-VLPAG microinjection of DM2 reduced the ongoing and tail flick-related activity of the nociceptive ON cells, whereas it increased the ongoing activity and reduced the tail flick-induced pause of the antinociceptive OFF cells, consistent with antinociception. Behavioural and electrophysiological effects were reproduced by intra-VLPAG microinjection of ethosuximide, a conventional T-type Ca(2+) channel blocker. Finally, DM2 administration did not produce any adverse cardiovascular effects as blood pressure and heart rate remained unchanged. In conclusion, DM2 plays an analgesic role in vivo and changes RVM cell activity, consistent with antinociception. These effects were even more potent than those elicited by ethosuximide treatments.


Subject(s)
Analgesics/pharmacology , Analgesics/therapeutic use , Medulla Oblongata/cytology , Neurons/drug effects , Pain/drug therapy , Pyridines/pharmacology , Action Potentials/drug effects , Animals , Anticonvulsants/pharmacology , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Ethosuximide/pharmacology , Formaldehyde/adverse effects , Heart Rate/drug effects , Male , Microinjections/methods , Neurons/physiology , Pain/chemically induced , Pain Measurement/methods , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiology , Rats , Rats, Wistar , Reaction Time/drug effects , Time Factors
13.
J Med Chem ; 52(12): 3794-800, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19459639

ABSTRACT

Although ketorolac is one of the most potent anti-inflammatory and analgesic drugs, its use has been strongly limited owing to the high incidence of adverse effects reported, particularly in the gastrointestinal tract. Using the prodrug approach, which allows the reduction of toxicological features of the parent drug without altering its pharmacological properties, we synthesized an orally administrable prodrug of ketorolac by means of its reversible conjugation to D-galactose (ketogal). In a single dose study, its pharmacokinetic profile was compared with that of ketorolac. Moreover, we found that this prodrug was able to maintain the anti-inflammatory and the analgesic activity of the drug without giving rise to gastric ulcer formation. Thus, these results indicate that ketogal is a highly effective and valid therapeutic alternative to ketorolac itself.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Galactose/chemistry , Ketorolac/pharmacology , Ketorolac/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Dose-Response Relationship, Drug , Edema/chemically induced , Hydrogen-Ion Concentration , Ketorolac/adverse effects , Ketorolac/chemistry , Mice , Molecular Conformation , Pain/chemically induced , Prodrugs/chemical synthesis , Prodrugs/chemistry , Stomach Ulcer/chemically induced , Time Factors
14.
J Biomed Mater Res A ; 87(1): 156-64, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18085645

ABSTRACT

Synthetic zeolites were studied in order to investigate their ability to encapsulate and to release drugs. In particular, a zeolite X and a zeolitic product obtained from a cocrystallization of zeolite X and zeolite A were examined. These materials were characterized by chemical analyses (ICP-AES), X-ray diffraction, nitrogen adsorption isotherm, scanning electron microscopy, laser diffraction, and infrared spectroscopy. Since ketoprofen was chosen as a model drug for the formulation of controlled-release dosage forms, it was encapsulated into these two types of synthetic zeolites by a soaking procedure. Drug-loaded matrices were then characterized for entrapped drug amount and thermogravimetric behavior. In both types of activated zeolites, the total amount of ketoprofen (800 mg) was encapsulated in 2 g of matrix. By using HPLC measurements, ketoprofen release studies were done at different pH conditions so as to mimick gastrointestinal fluids. The absence of release in acid conditions and a double phased release, at two different pH values (5 and 6.8), suggest that after activation these materials offer good potential for a modified release delivery system of ketoprofen.


Subject(s)
Drug Carriers/chemistry , Ketoprofen/chemistry , Zeolites/chemistry , Adsorption , Chemistry, Pharmaceutical/methods , Chromatography, High Pressure Liquid , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Drug Stability , Hydrogen-Ion Concentration , Ketoprofen/pharmacokinetics , Microscopy, Electron, Scanning , Nitrogen/chemistry , Particle Size , Spectroscopy, Near-Infrared , Thermogravimetry , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL