Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Cachexia Sarcopenia Muscle ; 15(2): 562-574, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38302863

ABSTRACT

BACKGROUND: Cancer-associated cachexia (CAC) is a debilitating syndrome associated with poor quality of life and reduced life expectancy of cancer patients. CAC is characterized by unintended body weight reduction due to muscle and adipose tissue loss. A major hallmark of CAC is systemic inflammation. Several non-steroidal anti-inflammatory drugs (NSAIDs) have been suggested for CAC treatment, yet no single medication has proven reliable. R-ketorolac (RK) is the R-enantiomer of a commonly used NSAID. The effect of RK on CAC has not yet been evaluated. METHODS: Ten- to 11-week-old mice were inoculated with C26 or CHX207 cancer cells or vehicle control (phosphate-buffered saline [PBS]). After cachexia onset, 2 mg/kg RK or PBS was administered daily by oral gavage. Body weight, food intake and tumour size were continuously measured. At study endpoints, blood was drawn, mice were sacrificed and tissues were excised. Immune cell abundance was analysed using a Cytek® Aurora spectral flow cytometer. Cyclooxygenase (COX) activity was determined in lung homogenates using a fluorometric kit. Muscle tissues were analysed for mRNA and protein expression by quantitative real-time PCR and western blotting analysis, respectively. Muscle fibre size was determined on histological slides after haematoxylin/eosin staining. RESULTS: Ten-day survival rate of C26-bearing animals was 10% while RK treatment resulted in a 100% survival rate (P = 0.0009). Chemotherapy resulted in a 10% survival rate 14 days after treatment initiation, but all mice survived upon co-medication with RK and cyclophosphamide (P = 0.0001). Increased survival was associated with a protection from body weight loss in C26 (-0.61 ± 1.82 vs. -4.48 ± 2.0 g, P = 0.0004) and CHX207 (-0.49 ± 0.33 vs. -2.49 ± 0.93 g, P = 0.0003) tumour-bearing mice treated with RK, compared with untreated mice. RK ameliorated musculus quadriceps (-1.7 ± 7.1% vs. -27.8 ± 8.3%, P = 0.0007) and gonadal white adipose tissue (-18.8 ± 49% vs. -69 ± 15.6%, P = 0.094) loss in tumour-bearing mice, compared with untreated mice. Mechanistically, RK reduced circulating interleukin-6 (IL-6) concentrations from 334 ± 151 to 164 ± 123 pg/mL (P = 0.047) in C26 and from 93 ± 39 to 35 ± 6 pg/mL (P = 0.0053) in CHX207 tumour-bearing mice. Moreover, RK protected mice from cancer-induced T-lymphopenia (+1.8 ± 42% vs. -49.2 ± 12.1% in treated vs. untreated mice, respectively). RK was ineffective in ameliorating CAC in thymus-deficient nude mice, indicating that the beneficial effect of RK depends on T-cells. CONCLUSIONS: RK improved T-lymphopenia and decreased systemic IL-6 concentrations, resulting in alleviation of cachexia and increased survival of cachexigenic tumour-bearing mice, even under chemotherapy and independent of COX inhibition. Considering its potential, we propose that the use of RK should be investigated in patients suffering from CAC.


Subject(s)
Lymphopenia , Neoplasms , Humans , Mice , Animals , Cachexia/drug therapy , Cachexia/etiology , Cachexia/metabolism , Ketorolac/metabolism , Ketorolac/pharmacology , Ketorolac/therapeutic use , Interleukin-6/metabolism , Mice, Nude , Quality of Life , Muscle, Skeletal/pathology , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/metabolism , Body Weight , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Lymphopenia/complications , Lymphopenia/drug therapy , Lymphopenia/pathology
2.
J Neuroimmunol ; 375: 578018, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36657373

ABSTRACT

Our objective was to determine whether (-)-Epicatechin administered alone or simultaneously with topical Ketorolac decreased the relative expression of GFAP and modulated the response of Nrf2 in a mouse model with induced hyperglycemia. We found that GFAP and Nrf2 decreased in the groups that received treatments alone or simultaneous during 8 weeks; even when the effect on the Nrf2 was not pronounced, it showed a higher concentration when GFAP decreased. Our results suggest a protective effect of Ketorolac and (-) - Epicatechin, which seem to limit the preclinical retinal damage caused by inflammation in hyperglycemia.


Subject(s)
Catechin , Hyperglycemia , Retinal Diseases , Animals , Mice , Catechin/pharmacology , Catechin/therapeutic use , Catechin/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Ketorolac/therapeutic use , Ketorolac/metabolism , Ketorolac/pharmacology , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Retina/metabolism
3.
Sci Rep ; 11(1): 17471, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471161

ABSTRACT

P21-activated kinases (PAKs) are serine/threonine protein kinase which have six different isoforms (PAK1-6). Of those, PAK1 is overexpressed in many cancers and considered to be a major chemotherapeutic target. Most of the developed PAK1 inhibitor drugs work as pan-PAK inhibitors and show undesirable toxicity due to having untargeted kinase inhibition activities. Selective PAK1 inhibitors are therefore highly desired and oncogenic drug hunters are trying to develop allosteric PAK1 inhibitors. We previously synthesized 1,2,3-triazolyl ester of ketorolac (15K) through click chemistry technique, which exhibits significant anti-cancer effects via inhibiting PAK1. Based on the selective anticancer effects of 15K against PAK1-dependent cancer cells, we hypothesize that it may act as an allosteric PAK1 inhibitor. In this study, computational analysis was done with 15K to explore its quantum chemical and thermodynamic properties, molecular interactions and binding stability with PAK1, physicochemical properties, ADMET, bioactivities, and druglikeness features. Molecular docking analysis demonstrates 15K as a potent allosteric ligand that strongly binds to a novel allosteric site of PAK1 (binding energy ranges - 8.6 to - 9.2 kcal/mol) and does not target other PAK isoforms; even 15K shows better interactions than another synthesized PAK1 inhibitor. Molecular dynamics simulation clearly supports the stable binding properties of 15K with PAK1 crystal. Density functional theory-based calculations reveal that it can be an active drug with high softness and moderate polarity, and ADMET predictions categorize it as a non-toxic drug as evidenced by in vitro studies with brine shrimp and fibroblast cells. Structure-activity relationship clarifies the role of ester bond and triazol moiety of 15K in establishing novel allosteric interactions. Our results summarize that 15K selectively inhibits PAK1 as an allosteric inhibitor and in turn shows anticancer effects without toxicity.


Subject(s)
Esters/chemistry , Ketorolac/metabolism , Models, Molecular , Oncogenes , Triazoles/chemistry , p21-Activated Kinases/chemistry , p21-Activated Kinases/metabolism , 3T3 Cells , Allosteric Regulation , Animals , Ketorolac/chemistry , Mice , Molecular Dynamics Simulation , Protein Conformation
4.
Cell Chem Biol ; 24(3): 281-292, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28238723

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used drugs in the world. While the role of NSAIDs as cyclooxygenase (COX) inhibitors is well established, other targets may contribute to anti-inflammation. Here we report caspases as a new pharmacological target for NSAID family drugs such as ibuprofen, naproxen, and ketorolac at physiologic concentrations both in vitro and in vivo. We characterize caspase activity in both in vitro and in cell culture, and combine computational modeling and biophysical analysis to determine the mechanism of action. We observe that inhibition of caspase catalysis reduces cell death and the generation of pro-inflammatory cytokines. Further, NSAID inhibition of caspases is COX independent, representing a new anti-inflammatory mechanism. This finding expands upon existing NSAID anti-inflammatory behaviors, with implications for patient safety and next-generation drug design.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Caspase Inhibitors/chemistry , Caspases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Caspase Inhibitors/metabolism , Caspase Inhibitors/pharmacology , Caspases/chemistry , Caspases, Initiator/chemistry , Caspases, Initiator/metabolism , Cell Line , Cell Survival/drug effects , High-Throughput Screening Assays , Humans , Ibuprofen/chemistry , Ibuprofen/metabolism , Ibuprofen/pharmacology , Inhibitory Concentration 50 , Ketorolac/chemistry , Ketorolac/metabolism , Ketorolac/pharmacology , Naproxen/chemistry , Naproxen/metabolism , Naproxen/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Substrate Specificity
5.
Eur J Med Chem ; 126: 270-276, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-27889630

ABSTRACT

An old anti-inflammatory/analgesic drug called Toradol is a racemic form of Ketorolac (50% R-form and 50% S-form) that blocks the oncogenic RAC-PAK1-COX-2 (cyclooxygenase-2) signaling, through the direct inhibition of RAC by the R-form and of COX-2 by the S-form, eventually down-regulating the production of prostaglandins. However, due to its COOH moiety which is clearly repulsive to negatively-charged phospholipid-based plasma membrane, its cell-permeability is rather poor (the IC50 against the growth of human cancer cells such as A549 is around 13 µM). In an attempt to boost its anti-cancer activity, hopefully by increasing its cell-permeability through abolishing the negative charge, yet keeping its water-solubility, here we synthesized a 1,2,3-triazolyl ester of Toradol through "Click Chemistry". The resultant water-soluble "azo" derivative called "15K" was found to be over 500 times more potent than Toradol with the IC50 around 24 nM against the PAK1-dependent growth of A549 cancer cells, inactivating PAK1 in cell culture with the apparent IC50 around 65 nM, and inhibiting COX-2 in vitro with the IC50 around 6 nM. Furthermore, the Click Chemistry boosts the anti-cancer activity of Ketorolac by 5000 times against the PAK1-independent growth of B16F10 melanoma cells. Using a multi-drug-resistant (MDR) cancer cell line (EMT6), we found that the esterization of Ketorolac boosts its cell-permeability by at least 10 folds. Thus, the Click Chemistry dramatically boosts the anti-cancer activity of Ketorolac, at least in three ways: increasing its cell-permeability, the anti-PAK1 activity of R-form and anti-COX-2 activity of S-form. The resultant "15K" is so far among the most potent PAK1-blockers, and therefore would be potentially useful for the therapy of many different PAK1-dependent diseases/disorders such as cancers.


Subject(s)
Esters/chemistry , Ketorolac/chemistry , Ketorolac/pharmacology , Triazoles/chemistry , p21-Activated Kinases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Click Chemistry , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Ketorolac/chemical synthesis , Ketorolac/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , p21-Activated Kinases/metabolism
6.
Drug Des Devel Ther ; 10: 1987-93, 2016.
Article in English | MEDLINE | ID: mdl-27382251

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most common type of medication used in the treatment of acute pain. Ketorolac trometamol (KT) is a nonnarcotic, peripherally acting nonsteroidal anti-inflammatory drug with analgesic effects comparable to certain opioids. OBJECTIVE: The aim of this study was to compare the efficacy of KT and naproxen (NA) in the treatment of acute low back pain (LBP) of moderate-to-severe intensity. PATIENTS AND METHODS: In this 10-day, Phase III, randomized, double-blind, double-dummy, noninferiority trial, participants with acute LBP of moderate-to-severe intensity as determined through a visual analog scale (VAS) were randomly assigned in a 1:1 ratio to receive sublingual KT 10 mg three times daily or oral NA 250 mg three times daily. From the second to the fifth day of treatment, if patient had VAS >40 mm, increased dosage to four times per day was allowed. The primary end point was the reduction in LBP as measured by VAS. We also performed a post hoc superiority analysis. RESULTS: KT was not inferior to NA for the reduction in LBP over 5 days of use as measured by VAS scores (P=0.608 for equality of variance; P=0.321 for equality of means) and by the Roland-Morris Disability Questionnaire (P=0.180 for equality of variance test; P=0.446 for equality of means) using 95% confidence intervals. The percentage of participants with improved pain relief 60 minutes after receiving the first dose was higher in the KT group (24.2%) than in the NA group (6.5%; P=0.049). The most common adverse effects were heartburn, nausea, and vomiting. CONCLUSION: KT is not inferior in efficacy and delivers faster pain relief than NA.


Subject(s)
Ketorolac/administration & dosage , Low Back Pain/drug therapy , Naproxen/administration & dosage , Tromethamine/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Double-Blind Method , Humans , Ketorolac/chemistry , Ketorolac/metabolism , Naproxen/chemistry , Naproxen/metabolism , Tromethamine/chemistry , Tromethamine/metabolism
7.
PLoS One ; 10(11): e0142182, 2015.
Article in English | MEDLINE | ID: mdl-26558612

ABSTRACT

Rho family GTPases (including Rac, Rho and Cdc42) collectively control cell proliferation, adhesion and migration and are of interest as functional therapeutic targets in numerous epithelial cancers. Based on high throughput screening of the Prestwick Chemical Library® and cheminformatics we identified the R-enantiomers of two approved drugs (naproxen and ketorolac) as inhibitors of Rac1 and Cdc42. The corresponding S-enantiomers are considered the active component in racemic drug formulations, acting as non-steroidal anti-inflammatory drugs (NSAIDs) with selective activity against cyclooxygenases. Here, we show that the S-enantiomers of naproxen and ketorolac are inactive against the GTPases. Additionally, more than twenty other NSAIDs lacked inhibitory action against the GTPases, establishing the selectivity of the two identified NSAIDs. R-naproxen was first identified as a lead compound and tested in parallel with its S-enantiomer and the non-chiral 6-methoxy-naphthalene acetic acid (active metabolite of nabumetone, another NSAID) as a structural series. Cheminformatics-based substructure analyses-using the rotationally constrained carboxylate in R-naproxen-led to identification of racemic [R/S] ketorolac as a suitable FDA-approved candidate. Cell based measurement of GTPase activity (in animal and human cell lines) demonstrated that the R-enantiomers specifically inhibit epidermal growth factor stimulated Rac1 and Cdc42 activation. The GTPase inhibitory effects of the R-enantiomers in cells largely mimic those of established Rac1 (NSC23766) and Cdc42 (CID2950007/ML141) specific inhibitors. Docking predicts that rotational constraints position the carboxylate moieties of the R-enantiomers to preferentially coordinate the magnesium ion, thereby destabilizing nucleotide binding to Rac1 and Cdc42. The S-enantiomers can be docked but are less favorably positioned in proximity to the magnesium. R-naproxen and R-ketorolac have potential for rapid translation and efficacy in the treatment of several epithelial cancer types on account of established human toxicity profiles and novel activities against Rho-family GTPases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ketorolac/pharmacology , Naproxen/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , HeLa Cells , Humans , Immunoblotting , Ketorolac/chemistry , Ketorolac/metabolism , Mice , Microscopy, Confocal , Molecular Docking Simulation , Molecular Structure , NIH 3T3 Cells , Naproxen/chemistry , Naproxen/metabolism , Protein Binding , Protein Structure, Tertiary , Stereoisomerism , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/metabolism
8.
Eur J Pharm Sci ; 70: 140-51, 2015 Apr 05.
Article in English | MEDLINE | ID: mdl-25676136

ABSTRACT

Migraine is a chronic disorder characterized by significant headache and various associated symptoms which worsen with exertion. Zolmitriptan approved for use in the acute treatment of migraine and related vascular headaches but are limited by high pain recurrence due to rapid drug elimination. Combinationalformulationof triptans and a nonsteroidal anti-inflammatory drug may provide a quicker and longer duration of relief from the subsequent pain during the attack. In this study, we formulate a Zolmitriptan (ZT) & ketorolac tromethamine (KT) loaded thermo reversible in-situ mucoadhesive intranasal gel (TMISG) formulation which gels at the nasal mucosal temperature and contains a bioadhesive polymer (Xyloglucan) that lengthens the residence time will enhance the bioavailability of the combinational drugs. This study uses Box-Behnken design for the first time to develop, optimize the TMISG and assess factors affecting the critical quality attributes. Histopathological study of the nasal mucosa suggested that the formulation was safe for nasal administration. The statistical difference in absolute bioavailability between oral and intranasal route suggested that intranasal route had almost 21% increases in bioavailability for ZT and for KT there was 16% increase over oral formulations. Optimized formulation would help mitigate migraine associated symptoms much better over the currently available formulations.


Subject(s)
Adhesives/administration & dosage , Drug Delivery Systems/methods , Ketorolac/administration & dosage , Migraine Disorders , Nasal Mucosa/drug effects , Oxazolidinones/administration & dosage , Tryptamines/administration & dosage , Adhesives/chemistry , Adhesives/metabolism , Administration, Intranasal , Animals , Disease Management , Gels , Ketorolac/chemistry , Ketorolac/metabolism , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Nasal Mucosa/metabolism , Oxazolidinones/chemistry , Oxazolidinones/metabolism , Sheep , Tryptamines/chemistry , Tryptamines/metabolism
9.
Drug Res (Stuttg) ; 65(8): 428-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25350079

ABSTRACT

The difference in tissue distribution of Ketorolac and its enantiomers were investigated in wistar rats. Separate high performance liquid chromatographic method was developed and validated for determination of Ketorolac and its enantiomers. Oyster BDS (150 × 4.6 mm id., 5 µm particle size) column was used for determination of concentration of Ketorolac. Ketorolac enantiomers were determined using Chiral-AGP column (100 × 4.0 mm I.D., particle size 5 µ, Chrom tech Ltd, Sweden). Detection was done at wavelength of 322 nm using an ultraviolet detector in the analytical system. Ketorolac enantiomers exhibit difference in their disposition in Wistar rats. In kidney, there was a significant difference in pharmacokinetic parameters. The Cmax was nearly 4 times and AUC 0-∞ was found to be more than double for S (-) Ketorolac than that of R (+) Ketorolac. MRT, Ke and t1/2 differ significantly in kidney. In liver, Cmax was found to be approximately 69% higher for S (-) Ketorolac compared to R (+) Ketorolac. AUC 0-∞ did not differ significantly for the enantiomers in liver. In liver, S (-) Ketorolac eliminated very fast in comparison to R (+) Ketorolac having t1/2 (one third) in comparison to R (+) Ketorolac. In lungs, there was no difference observed for Cmax and other parameters but AUC 0-∞ was found to be marginally higher for S (-) ketorolac.


Subject(s)
Ketorolac/metabolism , Tissue Distribution/physiology , Animals , Male , Rats , Rats, Wistar , Stereoisomerism
10.
Ukr Biochem J ; 86(3): 98-106, 2014.
Article in Ukrainian | MEDLINE | ID: mdl-25033559

ABSTRACT

Methods of fluorescent probing, spectrophotometry and microcalorimetry were applied to investigate the alterations in biophysical parameters of erythrocytes membranes, and specifically microviscosity, surface charge, molecular organization of lipid bilayer and lipid-protein interactions under conditions of acute pain syndrome produced by experimental chemical lesion. The distinctive features of non-opiod analgesics interactions and binding to the erythrocytes membranes of rats subjected to acute nociceptive pain accompanied with oxidative stress development were investigated. The abilities of analgesics under research, and namely paracetamol, aspirin, phenazone, ketorolac, pyrodazole, ketoprofenum, natrium mefenaminate, indometacin, nimesulide to make up physico-chemical complexes with lipoperoxidation modified erythrocytes surface and protein-lipid bilayer showed marked changes. The significance of oxidative damage of biophase under conditions of acute pain syndrome for analgesics effective pharmacodynamics and pharmacokinetics realization is under consideration.


Subject(s)
Acute Pain/prevention & control , Analgesics, Non-Narcotic/metabolism , Erythrocyte Membrane/metabolism , Lipid Bilayers/metabolism , Acetaminophen/metabolism , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Antipyrine/metabolism , Antipyrine/pharmacology , Aspirin/metabolism , Aspirin/pharmacology , Calorimetry , Cell Fractionation , Erythrocyte Membrane/chemistry , Fluorescent Dyes , Indomethacin/metabolism , Indomethacin/pharmacology , Ketoprofen/metabolism , Ketoprofen/pharmacology , Ketorolac/metabolism , Ketorolac/pharmacology , Lipid Bilayers/chemistry , Lipid Peroxidation/drug effects , Male , Orphenadrine/metabolism , Orphenadrine/pharmacology , Pain Measurement , Rats , Spectrometry, Fluorescence , Sulfonamides/metabolism , Sulfonamides/pharmacology
11.
J Pharm Sci ; 103(3): 974-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24481782

ABSTRACT

Different types of ketorolac ester prodrugs incorporating tert-butyl (KT), benzyl (KB), heptyl (KH), and diketorolac heptyl (DKH) promoieties were synthesized for the comparison of percutaneous penetration. The prodrugs were characterized according to their melting point, capacity factor, lipophilicity, solubility in 30% ethanol/buffer, enzymatic hydrolysis, in vitro skin permeation, hair follicle accumulation, and in vivo skin tolerance. Interactions between the prodrugs and esterases were predicted by molecular docking. Both equimolar suspensions and saturated solutions in 30% ethanol/pH 7.4 buffer were employed as the applied dose. All of the prodrugs exhibited a lower melting point than ketorolac. The lipophilicity increased in the following order: ketorolac < KT < KB < KH < DKH. The prodrugs were rapidly hydrolyzed to the parent drug in esterase medium, skin homogenate, and plasma, with KT and KB exhibiting higher degradation rates. KT exhibited the highest skin permeation, followed by KB. The flux of KT and KB exceeded that of ketorolac by 2.5-fold and twofold, respectively. KH and DKH did not improve ketorolac permeation but exhibited a sustained release behavior. KT and KH revealed selective absorption into follicles and a threefold greater follicular uptake compared with ketorolac. KB, KH, and DKH slightly but significantly increased transepidermal water loss (TEWL) after consecutive administration for 7 days, whereas ketorolac and KT exhibited no influence on TEWL. According to the experimental results, it can be concluded that an optimal balance between lipophilicity and aqueous solubility is important in the design of a successful prodrug. The acceptable skin tolerance for safe application is also an important consideration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Ketorolac/administration & dosage , Prodrugs/administration & dosage , Skin Absorption , Transdermal Patch , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Biotransformation , Esterases/chemistry , Esterases/metabolism , Esterification , Female , Hair Follicle/cytology , Hair Follicle/drug effects , Hair Follicle/metabolism , Humans , Hydrolysis , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Ketorolac/adverse effects , Ketorolac/chemistry , Ketorolac/metabolism , Mice , Mice, Nude , Molecular Docking Simulation , Prodrugs/adverse effects , Prodrugs/chemistry , Prodrugs/metabolism , Skin/cytology , Skin/drug effects , Skin/metabolism , Solubility , Sus scrofa , Tissue Distribution , Transition Temperature
12.
Eur J Pharm Biopharm ; 86(2): 219-26, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23791718

ABSTRACT

The objective of the study was to investigate the iontophoretic delivery kinetics of ketorolac (KT), a highly potent NSAID and peripherally-acting analgesic that is currently indicated to treat moderate to severe acute pain. It was envisaged that, depending on the amounts delivered, transdermal iontophoretic administration might have two distinct therapeutic applications: (i) more effective and faster local therapy with shorter onset times (e.g. to treat trauma-related pain/inflammation in muscle) or (ii) a non-parenteral, gastrointestinal tract sparing approach for systemic pain relief. The first part of the study investigated the effect of experimental conditions on KT iontophoresis using porcine and human skin in vitro. These results demonstrated that KT electrotransport was linearly dependent on current density - from 0.1875 to 0.5mA/cm(2) - (r(2)>0.99) and drug concentration - from 5 to 20mg/ml (r(2)>0.99). Iontophoretic permeation of KT from a 2% hydroxymethyl cellulose gel was comparable to that from an aqueous solution with equivalent drug loading (584.59±114.67 and 462.05±66.56µg/cm(2), respectively). Cumulative permeation (462.05±66.56 and 416.28±95.71µg/cm(2)) and steady state flux (106.72±11.70 and 94.28±15.47µg/cm(2)h), across porcine and human skin, were statistically equivalent confirming the validity of the model. Based on the results in vitro, it was decided to focus on topical rather than systemic applications of KT iontophoresis in vivo. Subsequent experiments, in male Wistar rats, investigated the local enhancement of KT delivery to muscle by iontophoresis. Drug biodistribution was assessed in skin, in the biceps femoris muscle beneath the site of iontophoresis ('treated muscle'; TM), in the contralateral muscle ('non-treated muscle'; NTM) and in plasma (P). Passive topical delivery and oral administration served as negative and positive controls, respectively. Iontophoretic administration for 30min was superior to passive topical delivery for 1h and resulted in statistically significant increases in KT levels in the skin (91.04±15.48 vs. 20.16±8.58µg/cm(2)), in the biceps femoris at the treatment site (TM; 6.74±3.80 vs.

Subject(s)
Ketorolac/administration & dosage , Ketorolac/metabolism , Muscles/drug effects , Muscles/metabolism , Administration, Cutaneous , Animals , Drug Delivery Systems/methods , Humans , Iontophoresis/methods , Kinetics , Male , Pain/drug therapy , Permeability , Rats , Rats, Wistar , Skin/drug effects , Skin/metabolism , Skin Absorption , Swine , Tissue Distribution
13.
J Mass Spectrom ; 47(7): 919-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22791260

ABSTRACT

In vivo metabolites of ketorolac (KTC) have been identified and characterized by using liquid chromatography positive ion electrospray ionization high resolution tandem mass spectrometry (LC/ESI-HR-MS/MS) in combination with online hydrogen/deuterium exchange (HDX) experiments. To identify in vivo metabolites, blood urine and feces samples were collected after oral administration of KTC to Sprague-Dawley rats. The samples were prepared using an optimized sample preparation approach involving protein precipitation and freeze liquid separation followed by solid-phase extraction and then subjected to LC/HR-MS/MS analysis. A total of 12 metabolites have been identified in urine samples including hydroxy and glucuronide metabolites, which are also observed in plasma samples. In feces, only O-sulfate metabolite and unchanged KTC are observed. The structures of metabolites were elucidated using LC-MS/MS and MS(n) experiments combined with accurate mass measurements. Online HDX experiments have been used to support the structural characterization of drug metabolites. The main phase I metabolites of KTC are hydroxylated and decarbonylated metabolites, which undergo subsequent phase II glucuronidation pathways.


Subject(s)
Chromatography, Liquid/methods , Ketorolac/chemistry , Ketorolac/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Deuterium Exchange Measurement , Feces/chemistry , Ketorolac/analysis , Ketorolac/pharmacokinetics , Male , Models, Molecular , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods
14.
Brain Res Mol Brain Res ; 125(1-2): 113-9, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15193428

ABSTRACT

Several lines of evidence have accumulated that release of excitatory amino acids, nitric oxide and prostaglandin E2 (PGE2) play a critical role in the development of peripheral tactile and thermal hypersensitivity in chronic inflammatory pain models. Synthesis of PGE2 is controlled by cyclooxygenase (COX), either the COX-1 or COX-2 isoform. COX-2 plays a central role in the inflammatory reactions. The relationship between central sensitization of a complete Freund's adjuvant (CFA) induced inflammation and expressions of COX-2 were assessed in a rat model of CFA injection induced inflammation. Moreover, the time course of analgesia and spinal COX-2 expression following intrathecal (IT) injection with a nonspecific COX inhibitor (ketorolac) and COX-2 inhibitor (celecoxib) were determined using Western blot and immunohistochemistry. COX-2 protein was slightly increased in the lumbosacral spinal cord at 24 h following subcutaneous injection of CFA in the plantar surface of the left hindpaw (p > 0.05). COX-1 was not detected in normal and CFA injection rats. Surprisingly, IT ketorolac or celecoxib significantly increased spinal COX-2 levels at 1 h post-IT injection (p < 0.05) both in inflamed and non-inflamed rats. Then, spinal COX-2 levels declined at 3 and 6 h post-IT injection. These results provide strong in vivo evidence that COX-2 activity but not level may play a central role in the Freund's adjuvant-induced inflammation. However, spinal COX-2 level was upregulated following IT ketorolac and celecoxib injection. These data implies that suppression of PGE2 activity may induce the expression of spinal COX-2 in Freund's adjuvant-induced pain model. Our study concludes that IT administration of COX-2 inhibitor or nonspecific COX inhibitor is associated with significant short-term increase in spinal COX-2 expression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Freund's Adjuvant/immunology , Inflammation/enzymology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Spinal Cord/enzymology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/administration & dosage , Hot Temperature , Hyperalgesia/metabolism , Injections, Spinal , Ketorolac/metabolism , Ketorolac/pharmacology , Male , Pain/drug therapy , Pain/metabolism , Pain Measurement , Pyrazoles , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/pathology , Sulfonamides/metabolism , Sulfonamides/pharmacology
15.
Anesth Analg ; 90(5): 1152-6, 2000 May.
Article in English | MEDLINE | ID: mdl-10781470

ABSTRACT

The management of severe pain may require "balanced analgesia," involving the use of analgesics with different modes of action. Clonidine, an alpha(2)-adrenoreceptor agonist produces analgesia by itself as well as when given with morphine and local anesthetics. Ketorolac is indicated for the management of moderately severe acute pain and causes analgesia equivalent to morphine. This study was designed to investigate whether the addition of ketorolac promotes antinociception produced by intrathecal administration of clonidine in male Sprague-Dawley rats. Intrathecal injection of clonidine (1-30 microg) induced a dose-dependent increase in antinociception as measured by the tail flick (TF) and hot plate tests. Ketorolac alone (150-600 microg) increased the antinociception by 50%-60% only in the TF test. Ketorolac (10 microg) decreased clonidine (10 microg)-induced antinociception from 69.1% +/- 7.8% to 23.5% +/- 1. 6% (P < 0.05) in the TF test and 35.7% +/- 4.7% to 4.5% +/- 0.1% (P < 0.05) maximum possible effect in the hot plate test. Ketorolac also antagonized the effect of 30 microg of clonidine. The opioid receptor antagonist naloxone antagonized the antinociceptive effect of clonidine and ketorolac, indicating the involvement of the opioid system in the antinociception produced by clonidine or ketorolac. However, neither clonidine nor ketorolac (10(-8) to 10(-3) M) inhibited the binding of specific ligands to mu-, delta-, and kappa-opioid receptors, indicating a lack of direct interaction of clonidine and ketorolac with opioid receptors. These results suggest that intrathecal injection of ketorolac antagonizes the antinociception produced by clonidine.


Subject(s)
Adrenergic alpha-Agonists/administration & dosage , Analgesia , Analgesics/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Clonidine/administration & dosage , Ketorolac/administration & dosage , Receptors, Opioid/metabolism , Adrenergic alpha-Agonists/metabolism , Analgesics/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Clonidine/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Injections, Spinal , Ketorolac/metabolism , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...