Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
2.
J Pharm Biomed Anal ; 225: 115220, 2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36610173

ABSTRACT

Ixazomib is the only orally active proteasome inhibitor used in clinical practice as an anticancer drug. The novel, rapid UHPLC-UV assay for ixazomib was developed and applied to the forced degradation study followed by HRMS identification of the main degradation products. Oxidative deboronation and hydrolysis of the amid bond were found to be the principal degradation pathways. The chemical standards of the main degradation products were prepared. The method was validated for the simultaneous assay of ixazomib and its degradation products within the concentration ranges of 2.50-100.00 µg/mL (ixazomib); 0.75-60.00 µg/mL (Impurity A and B) and 1.25-60.00 µg/mL (Impurity C). The stability study revealed that ixazomib in solution is: 1) relatively stable in neutral and acidic environments, 2) its decomposition is accelerated at higher pH, 3) it is sensitive to the effects of oxidants and light, and 4) the degradation of ixazomib follows the first-order kinetics under neutral, acidic, alkaline, and UV stress. Contrary, the solid substance of ixazomib citrate was relatively resistant to heat (70 °C), heat/humidity (70 °C/75 % RH), and UV irradiation for 24 h. This study presents the first MS-compatible UHPLC method for the quantification of ixazomib and its degradation products. Furthermore, it provides data about the inherent stability and kinetics of degradation of ixazomib in a solution that may be useful in further investigation of this drug, or the development of novel proteasome inhibitors based on the ixazomib structure.


Subject(s)
Antineoplastic Agents , Glycine , Chromatography, High Pressure Liquid/methods , Boron Compounds , Proteasome Inhibitors , Drug Stability , Hydrolysis , Oxidation-Reduction
4.
Pharmaceuticals (Basel) ; 15(5)2022 May 06.
Article in English | MEDLINE | ID: mdl-35631406

ABSTRACT

Antimicrobial drug resistance is currently one of the most critical health issues. Pathogens resistant to last-resort antibiotics are increasing, and very few effective antibacterial agents have been introduced in recent years. The promising drug candidates are often discontinued in the primary stages of the drug discovery pipeline due to their unspecific reactivity (PAINS), toxicity, insufficient stability, or low water solubility. In this work, we investigated a series of substituted N-oxazolyl- and N-thiazolylcarboxamides of various pyridinecarboxylic acids. Final compounds were tested against several microbial species. In general, oxazole-containing compounds showed high activity against mycobacteria, especially Mycobacterium tuberculosis (best MICH37Ra = 3.13 µg/mL), including the multidrug-resistant strains. Promising activities against various bacterial and fungal strains were also observed. None of the compounds was significantly cytotoxic against the HepG2 cell line. Experimental measurement of lipophilicity parameter log k'w and water solubility (log S) confirmed significantly (typically two orders in logarithmic scale) increased hydrophilicity/water solubility of oxazole derivatives in comparison with their thiazole isosteres. Mycobacterial ß-ketoacyl-acyl carrier protein synthase III (FabH) was suggested as a probable target by molecular docking and molecular dynamics simulations.

5.
Molecules ; 26(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918638

ABSTRACT

Gastrointestinal side effects of donepezil, including dyspepsia, nausea, vomiting or diarrhea, occur in 20-30% of patients. The pathogenesis of these dysmotility associated disorders has not been fully clarified yet. Pharmacokinetic parameters of donepezil and its active metabolite 6-O-desmethyldonepezil were investigated in experimental pigs with and without small intestinal injury induced by dextran sodium sulfate (DSS). Morphological features of this injury were evaluated by a video capsule endoscopy. The effect of a single and repeated doses of donepezil on gastric myoelectric activity was assessed. Both DSS-induced small intestinal injury and prolonged small intestinal transit time caused higher plasma concentrations of donepezil in experimental pigs. This has an important implication for clinical practice in humans, with a need to reduce doses of the drug if an underlying gastrointestinal disease is present. Donepezil had an undesirable impact on porcine myoelectric activity. This effect was further aggravated by DSS-induced small intestinal injury. These findings can explain donepezil-associated dyspepsia in humans.


Subject(s)
Donepezil/pharmacokinetics , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Indans/metabolism , Metabolome , Myoelectric Complex, Migrating , Piperidines/metabolism , Stomach/physiopathology , Animals , Capsule Endoscopy , Dextran Sulfate , Donepezil/chemistry , Donepezil/pharmacology , Female , Gastrointestinal Tract/drug effects , Metabolome/drug effects , Myoelectric Complex, Migrating/drug effects , Stomach/drug effects , Swine
6.
Int J Mol Sci ; 21(21)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33114215

ABSTRACT

Antidotes against organophosphates often possess physicochemical properties that mitigate their passage across the blood-brain barrier. Cucurbit[7]urils may be successfully used as a drug delivery system for bisquaternary oximes and improve central nervous system targeting. The main aim of these studies was to elucidate the relationship between cucurbit[7]uril, oxime K027, atropine, and paraoxon to define potential risks or advantages of this delivery system in a complex in vivo system. For this reason, in silico (molecular docking combined with umbrella sampling simulation) and in vivo (UHPLC-pharmacokinetics, toxicokinetics; acetylcholinesterase reactivation and functional observatory battery) methods were used. Based on our results, cucurbit[7]urils affect multiple factors in organophosphates poisoning and its therapy by (i) scavenging paraoxon and preventing free fraction of this toxin from entering the brain, (ii) enhancing the availability of atropine in the central nervous system and by (iii) increasing oxime passage into the brain. In conclusion, using cucurbit[7]urils with oximes might positively impact the overall treatment effectiveness and the benefits can outweigh the potential risks.


Subject(s)
Atropine/chemistry , Bridged-Ring Compounds/chemistry , Imidazoles/chemistry , Oximes/chemistry , Paraoxon/toxicity , Pyridinium Compounds/chemistry , Animals , Blood-Brain Barrier , Cholinesterase Reactivators/chemistry , Cholinesterase Reactivators/toxicity , Computer Simulation , Mice , Molecular Docking Simulation , Paraoxon/chemistry
7.
PLoS One ; 15(1): e0227781, 2020.
Article in English | MEDLINE | ID: mdl-31978146

ABSTRACT

BACKGROUND: Memantine, currently available for the treatment of Alzheimer's disease, is an uncompetitive antagonist of the N-methyl-D-aspartate type of glutamate receptors. Under normal physiologic conditions, these unstimulated receptor ion channels are blocked by magnesium ions, which are displaced after agonist-induced depolarization. In humans, memantine administration is associated with different gastrointestinal dysmotility side effects (vomiting, diarrhoea, constipation, motor-mediated abdominal pain), thus limiting its clinical use. Mechanism of these motility disorders has not been clarified yet. Pigs can be used in various preclinical experiments due to their relatively very similar gastrointestinal functions compared to humans. The aim of this study was to evaluate the impact of a single and repeated doses of memantine on porcine gastric myoelectric activity evaluated by means of electrogastrography (EGG). METHODS: Six adult female experimental pigs (Sus scrofa f. domestica, mean weight 41.7±5.0 kg) entered the study for two times. The first EGG was recorded after a single intragastric dose of memantine (20 mg). In the second part, EGG was accomplished after 7-day intragastric administration (20 mg per day). All EGG recordings were performed under general anaesthesia. Basal (15 minutes) and study recordings (120 minutes) were accomplished using an EGG stand (MMS, Enschede, the Netherlands). Running spectral analysis based on Fourier transform was used. Results were expressed as dominant frequency of gastric slow waves (DF) and power analysis (areas of amplitudes). RESULTS: Single dose of memantine significantly increased DF, from basic values (1.65±1.05 cycles per min.) to 2.86 cpm after 30 min. (p = 0.008), lasting till 75 min. (p = 0.014). Basal power (median 452; inter-quartile range 280-1312 µV^2) raised after 15 min. (median 827; IQR 224-2769; p = 0.386; NS), lasting next 30 min. Repetitively administrated memantine caused important gastric arrhythmia. Basal DF after single and repeated administration was not different, however, a DF increase in the second part was more prominent (up to 3.18±2.16 after 15 and 30 min., p<0.001). In comparison with a single dose, basal power was significantly higher after repetitively administrated memantine (median 3940; IQR 695-15023 µV^2; p<0.001). Next dose of 20 mg memantine in the second part induced a prominent drop of power after 15 min. (median 541; IQR 328-2280 µV^2; p<0.001), lasting till 120 min. (p<0.001). CONCLUSIONS: Both single and repeated doses of memantine increased DF. Severe gastric arrhythmia and long-lasting low power after repeated administration might explain possible gastric dysmotility side effects in the chronic use of memantine.


Subject(s)
Excitatory Amino Acid Antagonists/adverse effects , Gastrointestinal Diseases/chemically induced , Gastrointestinal Motility/drug effects , Memantine/adverse effects , Stomach/drug effects , Administration, Oral , Alzheimer Disease/drug therapy , Animals , Disease Models, Animal , Electromyography , Excitatory Amino Acid Antagonists/administration & dosage , Female , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/physiopathology , Gastrointestinal Motility/physiology , Humans , Memantine/administration & dosage , Stomach/physiopathology , Sus scrofa
9.
Toxicology ; 392: 1-10, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28941780

ABSTRACT

Novel dexrazoxane derivative JR-311 was prepared to investigate structure-activity relationships and mechanism(s) of protection against anthracycline cardiotoxicity. Its cardioprotective, antiproliferative, iron (Fe) chelation and inhibitory and/or depletory activities on topoisomerase IIbeta (TOP2B) were examined and compared with dexrazoxane. While in standard assay, JR-311 failed in both cardioprotection and depletion of TOP2B, its repeated administration to cell culture media led to depletion of TOP2B and significant protection of isolated rat neonatal ventricular cardiomyocytes from daunorubicin-induced damage. This effect was explained by a focused analytical investigation that revealed rapid JR-311 decomposition, resulting in negligible intracellular concentrations of the parent compound but high exposure of cells to the decomposition products, including Fe-chelating JR-H2. Although chemical instability is an obstacle for the development of JR-311, this study identified a novel dexrazoxane analogue with preserved pharmacodynamic properties, contributed to the investigation of structure-activity relationships and suggested that the cardioprotection of bis-dioxopiperazines is likely attributed to TOP2B activity of the parent compound rather than Fe chelation of their hydrolytic metabolites/degradation products. Moreover, this study highlights the importance of early stability testing during future development of novel dexrazoxane analogues.


Subject(s)
Cardiotonic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Dexrazoxane/pharmacology , Iron Chelating Agents/pharmacology , Animals , Animals, Newborn , Anthracyclines/toxicity , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cell Proliferation/drug effects , Cells, Cultured , Daunorubicin/toxicity , Dexrazoxane/analogs & derivatives , Diketopiperazines/pharmacology , Iron/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar , Structure-Activity Relationship
10.
J Pharm Biomed Anal ; 124: 365-373, 2016 May 30.
Article in English | MEDLINE | ID: mdl-26994320

ABSTRACT

This study addresses the lack of data on the stability of carfilzomib, a newly approved proteasome-inhibiting anticancer drug. A new stability-indicating UHPLC-UV method for analysis of carfilzomib was developed and validated within the concentrations of 10-250 µg/mL. The aforementioned method was utilized to evaluate the effects of forced degradation and to investigate the degradation kinetics, as well as to examine drug stability in a pharmaceutical formulation. A UHPLC-QTOF method was utilized to identify the principal degradation products. It was found that carfilzomib: (1) is stable at neutral and slightly acidic pH, but prone to degradation at both high and low pH; (2) is acceptably stable in the pharmaceutical formulation; but (3) is prone to oxidation and photodegradation. Carfilzomib degradation followed first-order kinetics. The decomposition products resulted from peptide bond hydrolysis, epoxide hydrolysis, hydrogen chloride addition, base-catalyzed Robinson-Gabriel reaction, tertiary amine oxidation and isomerization. Our results document, for the first time, the inherent stability of carfilzomib and provide information about the identity of its degradation products. These results highlight the stability issues that need to be kept in mind for handling and storage of carfilzomib.


Subject(s)
Chromatography, High Pressure Liquid/methods , Oligopeptides/analysis , Proteasome Inhibitors/analysis , Spectrophotometry, Ultraviolet/methods , Kinetics , Mass Spectrometry
11.
Oncotarget ; 6(40): 42411-28, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26623727

ABSTRACT

Di(2-pyridyl)ketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di(2-pyridyl)ketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) are novel, highly potent and selective anti-tumor and anti-metastatic drugs. Despite their structural similarity, these agents differ in their efficacy and toxicity in-vivo. Considering this, a comparison of their pharmacokinetic and pharmaco/toxico-dynamic properties was conducted to reveal if these factors are involved in their differential activity. Both compounds were administered to Wistar rats intravenously (2 mg/kg) and their metabolism and disposition were studied using UHPLC-MS/MS. The cytotoxicity of both thiosemicarbazones and their metabolites was also examined using MCF-7, HL-60 and HCT116 tumor cells and 3T3 fibroblasts and H9c2 cardiac myoblasts. Their intracellular iron-binding ability was characterized by the Calcein-AM assay and their iron mobilization efficacy was evaluated. In contrast to DpC, Dp44mT undergoes rapid demethylation in-vivo, which may be related to its markedly faster elimination (T1/2 = 1.7 h for Dp44mT vs. 10.7 h for DpC) and lower exposure. Incubation of these compounds with cancer cells or cardiac myoblasts did not result in any significant metabolism in-vitro. The metabolism of Dp44mT in-vivo resulted in decreased anti-cancer activity and toxicity. In conclusion, marked differences in the pharmacology of Dp44mT and DpC were observed and highlight the favorable pharmacokinetics of DpC for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Thiosemicarbazones/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Humans , Male , Rats , Rats, Wistar , Tandem Mass Spectrometry , Thiosemicarbazones/metabolism , Thiosemicarbazones/pharmacokinetics
12.
PLoS One ; 10(10): e0139929, 2015.
Article in English | MEDLINE | ID: mdl-26460540

ABSTRACT

Cancer cells have a high iron requirement and many experimental studies, as well as clinical trials, have demonstrated that iron chelators are potential anti-cancer agents. The ligand, 2-benzoylpyridine 4-ethyl-3-thiosemicarbazone (Bp4eT), demonstrates both potent anti-neoplastic and anti-retroviral properties. In this study, Bp4eT and its recently identified amidrazone and semicarbazone metabolites were examined and compared with respect to their anti-proliferative activity towards cancer cells (HL-60 human promyelocytic leukemia, MCF-7 human breast adenocarcinoma, HCT116 human colon carcinoma and A549 human lung adenocarcinoma), non-cancerous cells (H9c2 neonatal rat-derived cardiomyoblasts and 3T3 mouse embryo fibroblasts) and their interaction with intracellular iron pools. Bp4eT was demonstrated to be a highly potent and selective anti-neoplastic agent that induces S phase cell cycle arrest, mitochondrial depolarization and apoptosis in MCF-7 cells. Both semicarbazone and amidrazone metabolites showed at least a 300-fold decrease in cytotoxic activity than Bp4eT towards both cancer and normal cell lines. The metabolites also lost the ability to: (1) promote the redox cycling of iron; (2) bind and mobilize iron from labile intracellular pools; and (3) prevent 59Fe uptake from 59Fe-labeled transferrin by MCF-7 cells. Hence, this study demonstrates that the highly active ligand, Bp4eT, is metabolized to non-toxic and pharmacologically inactive analogs, which most likely contribute to its favorable pharmacological profile. These findings are important for the further development of this drug candidate and contribute to the understanding of the structure-activity relationships of these agents.


Subject(s)
Antineoplastic Agents/pharmacology , Iron Chelating Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Thiosemicarbazones/pharmacology , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Iron/chemistry , Iron/metabolism , Iron Chelating Agents/chemistry , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Semicarbazones/chemistry , Semicarbazones/metabolism , Semicarbazones/pharmacology , Semicarbazones/toxicity , Thiosemicarbazones/chemistry , Thiosemicarbazones/metabolism , Thiosemicarbazones/toxicity
13.
Biomed Chromatogr ; 28(5): 621-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24254882

ABSTRACT

Novel thiosemicarbazone metal chelators are extensively studied anti-cancer agents with marked and selective activity against a wide variety of cancer cells, as well as human tumor xenografts in mice. This study describes the first validated LC-MS/MS method for the simultaneous quantification of 2-benzoylpyridine 4-ethyl-3-thiosemicarbazone (Bp4eT) and its main metabolites (E/Z isomers of the semicarbazone structure, M1-E and M1-Z, and the amidrazone metabolite, M2) in plasma. Separation was achieved using a C18 column with ammonium formate/acetonitrile mixture as the mobile phase. Plasma samples were treated using solid-phase extraction on 96-well plates. This method was validated over the concentration range of 0.18-2.80 µM for Bp4eT, 0.02-0.37 µM for both M1-E and M1-Z, and 0.10-1.60 µM for M2. This methodology was applied to the analysis of samples from in vivo experiments, allowing for the concentration-time profile to be simultaneously assessed for the parent drug and its metabolites. The current study addresses the lack of knowledge regarding the quantitative analysis of thiosemicarbazone anti-cancer drugs and their metabolites in plasma and provides the first pharmacokinetic data on a lead compound of this class.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Thiosemicarbazones/blood , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Male , Pilot Projects , Rats , Rats, Wistar , Thiosemicarbazones/metabolism , Thiosemicarbazones/pharmacokinetics
14.
Anal Bioanal Chem ; 403(1): 309-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22349326

ABSTRACT

The iron chelator, 2-benzoylpyridine-4-ethyl-3-thiosemicarbazone (Bp4eT), was identified as a lead compound of the 2-benzoylpyridine thiosemicarbazone series, which were designed as potential anti-cancer agents. This ligand has been shown to possess potent anti-proliferative activity with a highly selective mechanism of action. However, further progress in the development of this compound requires data regarding its metabolism in mammals. The aim of this study was to identify the main in vitro and in vivo phase I metabolites of Bp4eT using liquid chromatography tandem mass spectrometry (LC-MS/MS). Two metabolites were detected after incubation of this drug with rat and human liver microsomal fractions. Based on LC-MS(n) analysis, the metabolites were demonstrated to be 2-benzoylpyridine-4-ethyl-3-semicarbazone and N (3)-ethyl-N (1)-[phenyl(pyridin-2-yl)methylene]formamidrazone, with both resulting from the oxidation of the thiocarbonyl group. The identity of these metabolites was further shown by LC-MS/MS analysis of these latter compounds which were prepared by oxidation of Bp4eT with hydrogen peroxide and their structures confirmed by nuclear magnetic resonance and infrared spectra. Both the semicarbazone and the amidrazone metabolites were detected in plasma, urine, and feces after i.v. administration of Bp4eT to rats. In addition, another metabolite that could correspond to hydroxylated amidrazone was found in vivo. Thus, oxidative pathways play a major role in the phase I metabolism of this promising anti-tumor agent. The outcomes of this study will be further utilized for: (1) the development and validation of the analytical method for the quantification of Bp4eT and its metabolites in biological materials; (2) to design pharmacokinetic experiments; and to (3) evaluate the potential contribution of the individual metabolites to the pharmacodynamics/toxico-dynamics of this novel anti-proliferative agent.


Subject(s)
Antineoplastic Agents/metabolism , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Thiosemicarbazones/metabolism , Animals , Humans , In Vitro Techniques , Magnetic Resonance Spectroscopy , Male , Rats , Solid Phase Extraction , Spectrophotometry, Infrared
SELECTION OF CITATIONS
SEARCH DETAIL