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










Publication year range
1.
Arch Pharm (Weinheim) ; : e2400171, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710636

ABSTRACT

This study presents an exploration of the chemical space around derivatives of 3-benzamidopyrazine-2-carboxamides, previously identified as potent antimycobacterial compounds with predicted binding to mycobacterial prolyl-transfer RNA synthetase. New urea derivatives (Series-1) were generally inactive, probably due to their preference for cis-trans conformation (confirmed by density functional theory calculations and experimentally by nuclear overhauser effect spectroscopy NMR). Series-2 (3-benzamidopyrazine-2-carboxamides with disubstituted benzene ring) demonstrated that substituents larger than fluorine are not tolerated in the ortho position of the benzene ring. This series brought two new compounds (21: R = 2-F, 4-Cl and 22: R = 2-F, 4-Br) with in vitro activity against Mycobacterium tuberculosis H37Rv as well as multidrug-resistant clinical isolates, with minimum inhibitory concentration ranging from 6.25 to 25 µg/mL. The lactone-type derivatives 4H-pyrazino[2,3-d][1,3]oxazin-4-ones (Series-3) were inactive, but solvent stability studies of compound 29 indicated that they might be developed to usable lactone prodrugs of inhibitors of mycobacterial aspartate decarboxylase (PanD).

2.
Toxicol Sci ; 198(2): 288-302, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38290791

ABSTRACT

Anthracyclines, such as doxorubicin (adriamycin), daunorubicin, or epirubicin, rank among the most effective agents in classical anticancer chemotherapy. However, cardiotoxicity remains the main limitation of their clinical use. Topoisomerase IIß has recently been identified as a plausible target of anthracyclines in cardiomyocytes. We examined the putative topoisomerase IIß selective agent XK469 as a potential cardioprotective and designed several new analogs. In our experiments, XK469 inhibited both topoisomerase isoforms (α and ß) and did not induce topoisomerase II covalent complexes in isolated cardiomyocytes and HL-60, but induced proteasomal degradation of topoisomerase II in these cell types. The cardioprotective potential of XK469 was studied on rat neonatal cardiomyocytes, where dexrazoxane (ICRF-187), the only clinically approved cardioprotective, was effective. Initially, XK469 prevented daunorubicin-induced toxicity and p53 phosphorylation in cardiomyocytes. However, it only partially prevented the phosphorylation of H2AX and did not affect DNA damage measured by Comet Assay. It also did not compromise the daunorubicin antiproliferative effect in HL-60 leukemic cells. When administered to rabbits to evaluate its cardioprotective potential in vivo, XK469 failed to prevent the daunorubicin-induced cardiac toxicity in either acute or chronic settings. In the following in vitro analysis, we found that prolonged and continuous exposure of rat neonatal cardiomyocytes to XK469 led to significant toxicity. In conclusion, this study provides important evidence on the effects of XK469 and its combination with daunorubicin in clinically relevant doses in cardiomyocytes. Despite its promising characteristics, long-term treatments and in vivo experiments have not confirmed its cardioprotective potential.


Subject(s)
Anthracyclines , Quinoxalines , Topoisomerase II Inhibitors , Rats , Animals , Rabbits , Topoisomerase II Inhibitors/toxicity , Topoisomerase II Inhibitors/therapeutic use , Anthracyclines/toxicity , Anthracyclines/therapeutic use , Cardiotoxicity , Daunorubicin/toxicity , Daunorubicin/therapeutic use , Doxorubicin/toxicity , Antibiotics, Antineoplastic/toxicity , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/therapeutic use , DNA Damage
3.
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.
J Pharm Biomed Anal ; 210: 114549, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-34998075

ABSTRACT

Breast milk analysis provides useful information about acute newborn exposure to harmful substances, such as psychoactive drugs abused by a nursing mother. Since breast milk represents a complex matrix with large amounts of interfering compounds, a comprehensive sample pre-treatment is necessary. This work focuses on determination of amphetamines and synthetic cathinones in human breast milk by microextraction techniques (liquid-phase microextraction and electromembrane extraction), and their comparison to more conventional treatment methods (protein precipitation, liquid-liquid extraction, and salting-out assisted liquid-liquid extraction). The aim of this work was to optimize and validate all the extraction procedures and thoroughly assess their advantages and disadvantages with special regard to their routine clinical use. The applicability of the extractions was further verified by the analysis of six real samples collected from breastfeeding mothers suspected of amphetamine abuse. The membrane microextraction techniques turned out to be the most advantageous as they required low amounts of organic solvents but still provided efficient sample clean-up, excellent quantification limit (0.5 ng mL-1), and good recovery (81-91% and 40-89% for electromembrane extraction and liquid-phase microextraction, respectively). The traditional liquid-liquid extraction as well as the salting-out assisted liquid-liquid extraction showed comparable recoveries (41-85% and 63-88%, respectively), but higher quantification limits (2.5 ng mL-1 and 5 ng mL-1, respectively). Moreover, these methods required multiple operating steps and were time consuming. Protein precipitation was fast and simple, but it demonstrated poor sample clean-up, low recovery (56-58%) and high quantification limit (5 ng mL-1). Based on the overall results, microextraction methods can be considered promising candidates, even for routine laboratory use.


Subject(s)
Liquid Phase Microextraction , Milk, Human , Amphetamines , Female , Humans , Infant, Newborn , Limit of Detection , Liquid-Liquid Extraction , Solvents
5.
Circ Heart Fail ; 14(11): e008209, 2021 11.
Article in English | MEDLINE | ID: mdl-34551586

ABSTRACT

BACKGROUND: Anthracycline-induced heart failure has been traditionally attributed to direct iron-catalyzed oxidative damage. Dexrazoxane (DEX)-the only drug approved for its prevention-has been believed to protect the heart via its iron-chelating metabolite ADR-925. However, direct evidence is lacking, and recently proposed TOP2B (topoisomerase II beta) hypothesis challenged the original concept. METHODS: Pharmacokinetically guided study of the cardioprotective effects of clinically used DEX and its chelating metabolite ADR-925 (administered exogenously) was performed together with mechanistic experiments. The cardiotoxicity was induced by daunorubicin in neonatal ventricular cardiomyocytes in vitro and in a chronic rabbit model in vivo (n=50). RESULTS: Intracellular concentrations of ADR-925 in neonatal ventricular cardiomyocytes and rabbit hearts after treatment with exogenous ADR-925 were similar or exceeded those observed after treatment with the parent DEX. However, ADR-925 did not protect neonatal ventricular cardiomyocytes against anthracycline toxicity, whereas DEX exhibited significant protective effects (10-100 µmol/L; P<0.001). Unlike DEX, ADR-925 also had no significant impact on daunorubicin-induced mortality, blood congestion, and biochemical and functional markers of cardiac dysfunction in vivo (eg, end point left ventricular fractional shortening was 32.3±14.7%, 33.5±4.8%, 42.7±1.0%, and 41.5±1.1% for the daunorubicin, ADR-925 [120 mg/kg]+daunorubicin, DEX [60 mg/kg]+daunorubicin, and control groups, respectively; P<0.05). DEX, but not ADR-925, inhibited and depleted TOP2B and prevented daunorubicin-induced genotoxic damage. TOP2B dependency of the cardioprotective effects was probed and supported by experiments with diastereomers of a new DEX derivative. CONCLUSIONS: This study strongly supports a new mechanistic paradigm that attributes clinically effective cardioprotection against anthracycline cardiotoxicity to interactions with TOP2B but not metal chelation and protection against direct oxidative damage.


Subject(s)
Anthracyclines/pharmacology , Cardiotoxicity/prevention & control , Dexrazoxane/pharmacology , Heart Failure/drug therapy , Topoisomerase II Inhibitors/metabolism , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacology , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , DNA Topoisomerases, Type II/adverse effects , DNA Topoisomerases, Type II/metabolism , Daunorubicin/metabolism , Daunorubicin/pharmacology , Dexrazoxane/adverse effects , Heart Diseases/drug therapy , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects
6.
Anal Chim Acta ; 1177: 338742, 2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34482886

ABSTRACT

The reliable analysis of various compounds from tissue requires a tedious sample preparation. The sample pretreatment usually involves proper homogenization that facilitates extraction of target analytes, followed by an appropriate sample clean-up preventing matrix effects. Electromembrane extraction (EME) seems to have a significant potential to streamline the whole procedure. In this study, the applicability of EME for direct isolation of analytes from animal tissues was investigated for the first time. Extraction conditions were systematically optimized to isolate model analytes (daunorubicin and its metabolite daunorubicinol) from various tissues (myocardium, skeletal muscle and liver) coming from a pharmacokinetic study in rabbits. The relative recoveries of daunorubicin and its metabolite in all tissues, determined by the UHPLC-MS/MS method, were higher than 66 and 75%, respectively. Considerably low matrix effects (0 ± 8% with CV lower than 6%) and negligible content of phospholipids detected in EME extracts demonstrate the exceptional effectiveness of this microextraction approach in purification of tissue samples. The difference in the concentrations of the analytes determined after EME and reference liquid-liquid extraction of real tissue samples was lower than 12%, which further emphasized the trustworthiness of EME. Moreover, the considerable time reduction needed for sample treatment in case of EME must be emphasized. This study proved that EME is a simple, effective and reliable microextraction technique capable of direct extraction of the analytes from pulverized tissues without the need for an additional homogenization or purification step.


Subject(s)
Pharmaceutical Preparations , Tandem Mass Spectrometry , Animals , Membranes, Artificial , Phospholipids , Rabbits
7.
Clin Sci (Lond) ; 135(15): 1897-1914, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34318878

ABSTRACT

The anthracycline (ANT) anticancer drugs such as doxorubicin or daunorubicin (DAU) can cause serious myocardial injury and chronic cardiac dysfunction in cancer survivors. A bisdioxopiperazine agent dexrazoxane (DEX) has been developed as a cardioprotective drug to prevent these adverse events, but it is uncertain whether it is the best representative of the class. The present study used a rabbit model of chronic ANT cardiotoxicity to examine another bisdioxopiperazine compound called GK-667 (meso-(butane-2,3-diylbis(2,6-dioxopiperazine-4,1-diyl))bis(methylene)-bis(2-aminoacetate) hydrochloride), a water-soluble prodrug of ICRF-193 (meso-4,4'-(butan-2,3-diyl)bis(piperazine-2,6-dione)), as a potential cardioprotectant. The cardiotoxicity was induced by DAU (3 mg/kg, intravenously, weekly, 10 weeks), and GK-667 (1 or 5 mg/kg, intravenously) was administered before each DAU dose. The treatment with GK-667 was well tolerated and provided full protection against DAU-induced mortality and left ventricular (LV) dysfunction (determined by echocardiography and LV catheterization). Markers of cardiac damage/dysfunction revealed minor cardiac damage in the group co-treated with GK-667 in the lower dose, whereas almost full protection was achieved with the higher dose. This was associated with similar prevention of DAU-induced dysregulation of redox and calcium homeostasis proteins. GK-667 dose-dependently prevented tumor suppressor p53 (p53)-mediated DNA damage response in the LV myocardium not only in the chronic experiment but also after single DAU administration. These effects appear essential for cardioprotection, presumably because of the topoisomerase IIß (TOP2B) inhibition provided by its active metabolite ICRF-193. In addition, GK-667 administration did not alter the plasma pharmacokinetics of DAU and its main metabolite daunorubicinol (DAUol) in rabbits in vivo. Hence, GK-667 merits further investigation as a promising drug candidate for cardioprotection against chronic ANT cardiotoxicity.


Subject(s)
Cardiomyopathies/prevention & control , DNA Damage , Diketopiperazines/pharmacology , Myocytes, Cardiac/drug effects , Prodrugs/pharmacology , Topoisomerase II Inhibitors/pharmacology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Cardiotoxicity , Chronic Disease , Daunorubicin , Disease Models, Animal , Fibrosis , HL-60 Cells , Humans , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rabbits , Tumor Suppressor Protein p53/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
8.
Sci Rep ; 11(1): 4456, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627707

ABSTRACT

The bisdioxopiperazine topoisomerase IIß inhibitor ICRF-193 has been previously identified as a more potent analog of dexrazoxane (ICRF-187), a drug used in clinical practice against anthracycline cardiotoxicity. However, the poor aqueous solubility of ICRF-193 has precluded its further in vivo development as a cardioprotective agent. To overcome this issue, water-soluble prodrugs of ICRF-193 were prepared, their abilities to release ICRF-193 were investigated using a novel UHPLC-MS/MS assay, and their cytoprotective effects against anthracycline cardiotoxicity were tested in vitro in neonatal ventricular cardiomyocytes (NVCMs). Based on the obtained results, the bis(2-aminoacetoxymethyl)-type prodrug GK-667 was selected for advanced investigations due to its straightforward synthesis, sufficient solubility, low cytotoxicity and favorable ICRF-193 release. Upon administration of GK-667 to NVCMs, the released ICRF-193 penetrated well into the cells, reached sufficient intracellular concentrations and provided effective cytoprotection against anthracycline toxicity. The pharmacokinetics of the prodrug, ICRF-193 and its rings-opened metabolite was estimated in vivo after administration of GK-667 to rabbits. The plasma concentrations of ICRF-193 reached were found to be adequate to achieve cardioprotective effects in vivo. Hence, GK-667 was demonstrated to be a pharmaceutically acceptable prodrug of ICRF-193 and a promising drug candidate for further evaluation as a potential cardioprotectant against chronic anthracycline toxicity.


Subject(s)
Anthracyclines/adverse effects , Cardiotonic Agents/pharmacology , Cardiotoxicity/drug therapy , DNA Topoisomerases, Type II/metabolism , Diketopiperazines/pharmacology , Piperazine/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cardiotonic Agents/chemistry , Cardiotoxicity/metabolism , Dexrazoxane/chemistry , Dexrazoxane/pharmacology , Diketopiperazines/chemistry , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Piperazine/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Rabbits , Razoxane/chemistry , Razoxane/pharmacology , Topoisomerase II Inhibitors/chemistry , Water/chemistry
9.
Talanta ; 223(Pt 2): 121748, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33298272

ABSTRACT

Electromembrane extraction (EME) of the polar zwitterionic drugs, anthracyclines (ANT, doxorubicin, daunorubicin and its metabolite daunorubicinol), from rabbit plasma was investigated. The optimized EME was compared to conventional sample pretreatment techniques such as protein precipitation (PP) and liquid-liquid extraction (LLE), mainly in terms of extraction reliability, recovery and matrix effect. In addition, phospholipids profile in the individual extracts was evaluated. The extracted samples were analyzed using UHPLC-MS/MS with electrospray ionization in positive ion mode. The method was validated within the concentration range of 0.25-1000 ng/mL for all tested ANT. Compared with PP and LLE, the EME provided high extraction recovery (more than 80% for all ANT) and excellent sample clean-up (matrix effect were 100 ± 10% with RSD values lower than 4% for all ANT). Furthermore, only negligible amounts of phospholipids were detected in the EME samples. Finally, practical applicability of EME was proved by analysis of plasma samples taken from a pilot in vivo study in rabbits. Consistent results were obtained when using both EME and LLE to extract the plasma prior to the analysis, which further confirmed high reliability of EME. This study clearly showed that EME is a simple, rapid, repeatable technique for extraction of ANT from plasma and it is an up to date alternative to routine conventional extraction techniques.


Subject(s)
Pharmaceutical Preparations , Tandem Mass Spectrometry , Animals , Anthracyclines , Membranes, Artificial , Rabbits , Reproducibility of Results
10.
Talanta ; 222: 121652, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33167277

ABSTRACT

Boron cluster compounds are extensively studied due to their possible use in medicinal chemistry, mainly in the boron neutron capture anticancer therapy and as new innovative pharmacophores. Concerning this research, the chiral separations of exceptionally stable anionic 7,8-dicarba-nido-undecaborate(1-) and metal bis(dicarbollide(1-) derivatives with asymmetric substitutions remain the unsolved challenge of the chiral chromatography nowadays. Although the successful enantioseparation of some anionic 7,8-dicarba-nido-undecaborate(1-) ion derivatives were achieved in CZE with native ß-cyclodextrins, it has not been observed with HPLC, yet. This study aimed to systematically investigate the enantioseparation of selected compounds in HPLC using native ß-cyclodextrin and brominated ß-cyclodextrin. The findings revealed positively charged strong adsorption sites on a stationary phase, identified as the cationic metal impurities in the silica-gel backbone. All the anionic species under the study were at least partially enantioseparated when a chelating agent blocked these cationic sites. Consequently, the first-ever HPLC enantioseparations of the 7,8-dicarba-nido-undecaborates(1-) were achieved. The brominated ß-cyclodextrin seemed to be a better chiral selector for separation of these species, whereas the native ß-cyclodextrin separated the anionic cobalt bis(dicarbollide(1-). The results of this study bring new information concerning the chiral separation of anionic boron clusters and might be used in the chiral method development process on other chiral selectors. Furthermore, the possibility of chiral separation of these species could influence the ongoing research areas of anionic boron clusters.


Subject(s)
Boron Compounds , Cyclodextrins , Anions , Cations , Chromatography, High Pressure Liquid , Stereoisomerism
11.
J Pharmacol Exp Ther ; 373(3): 402-415, 2020 06.
Article in English | MEDLINE | ID: mdl-32253261

ABSTRACT

Bisdioxopiperazine agent dexrazoxane (ICRF-187) has been the only effective and approved drug for prevention of chronic anthracycline cardiotoxicity. However, the structure-activity relationships (SARs) of its cardioprotective effects remain obscure owing to limited investigation of its derivatives/analogs and uncertainties about its mechanism of action. To fill these knowledge gaps, we tested the hypothesis that dexrazoxane derivatives exert cardioprotection via metal chelation and/or modulation of topoisomerase IIß (Top2B) activity in chronic anthracycline cardiotoxicity. Dexrazoxane was alkylated in positions that should not interfere with the metal-chelating mechanism of cardioprotective action; that is, on dioxopiperazine imides or directly on the dioxopiperazine ring. The protective effects of these agents were assessed in vitro in neonatal cardiomyocytes. All studied modifications of dexrazoxane molecule, including simple methylation, were found to abolish the cardioprotective effects. Because this challenged the prevailing mechanistic concept and previously reported data, the two closest derivatives [(±)-4,4'-(propane-1,2-diyl)bis(1-methylpiperazine-2,6-dione) and 4-(2-(3,5-dioxopiperazin-1-yl)ethyl)-3-methylpiperazine-2,6-dione] were thoroughly scrutinized in vivo using a rabbit model of chronic anthracycline cardiotoxicity. In contrast to dexrazoxane, both compounds failed to protect the heart, as demonstrated by mortality, cardiac dysfunction, and myocardial damage parameters, although the pharmacokinetics and metal-chelating properties of their metabolites were comparable to those of dexrazoxane. The loss of cardiac protection was shown to correlate with their abated potential to inhibit and deplete Top2B both in vitro and in vivo. These findings suggest a very tight SAR between bisdioxopiperazine derivatives and their cardioprotective effects and support Top2B as a pivotal upstream druggable target for effective cardioprotection against anthracycline cardiotoxicity. SIGNIFICANCE STATEMENT: This study has revealed the previously unexpected tight structure-activity relationships of cardioprotective effects in derivatives of dexrazoxane, which is the only drug approved for the prevention of cardiomyopathy and heart failure induced by anthracycline anticancer drugs. The data presented in this study also strongly argue against the importance of metal-chelating mechanisms for the induction of this effect and support the viability of topoisomerase IIß as an upstream druggable target for effective and clinically translatable cardioprotection.


Subject(s)
Anthracyclines/adverse effects , Cardiotoxicity/drug therapy , DNA Topoisomerases, Type II/metabolism , Dexrazoxane/pharmacology , Heart/drug effects , Protective Agents/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/metabolism , Cell Line, Tumor , HL-60 Cells , Humans , Male , Models, Animal , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rabbits , Rats , Rats, Wistar , Structure-Activity Relationship
12.
Sci Rep ; 9(1): 4524, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872754

ABSTRACT

Sobuzoxane (MST-16) is an approved anticancer agent, a pro-drug of bisdioxopiperazine analog ICRF-154. Due to the structural similarity of ICRF-154 to dexrazoxane (ICRF-187), MST-16 deserves attention as a cardioprotective drug. This study presents for the first time UHPLC-MS/MS assay of MST-16, ICRF-154 and its metabolite (EDTA-diamide) in cell culture medium, buffer, plasma and cardiac cells and provides data on MST-16 bioactivation under conditions relevant to investigation of cardioprotection of this drug. The analysis of these compounds that differ considerably in their lipophilicity was achieved on the Zorbax SB-Aq column using a mixture of aqueous ammonium formate and methanol as a mobile phase. The biological samples were either diluted or precipitated with methanol, which was followed by acidification for the assay of MST-16. The method was validated for determination of all compounds in the biological materials. The application of the method for analysis of samples from in vitro experiments provided important findings, namely, that (1) MST-16 is quickly decomposed in biological environments, (2) the cardiac cells actively metabolize MST-16, and (3) MST-16 readily penetrates into the cardiac cells and is converted into ICRF-154 and EDTA-diamide. These data are useful for the in-depth examination of the cardioprotective potential of this drug.


Subject(s)
Antineoplastic Agents/analysis , Edetic Acid/chemistry , Piperazines/analysis , Razoxane/analogs & derivatives , Animals , Antineoplastic Agents/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar , Razoxane/chemistry , Razoxane/metabolism , Tandem Mass Spectrometry
13.
Anal Bioanal Chem ; 411(11): 2383-2394, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30820631

ABSTRACT

Solid-phase microextraction (SPME) is an alternative method to dialysis and ultrafiltration for the determination of plasma protein binding (PPB) of drugs. It is particularly advantageous for complicated analytes where standard methods are not applicable. Di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) is a lead compound of novel thiosemicarbazone anti-cancer drugs, which entered clinical trials in 2016. However, this agent exhibited non-specific binding on filtration membranes and had intrinsic chelation activity, which precluded standard PPB methods. In this study, using a simple and fast procedure, we prepared novel SPME fibers for extraction of DpC based on a metal-free, silicon string support, covered with C18 sorbent. Reproducibility of the preparation process was demonstrated by the percent relative standard deviation (RSD) of ≤ 9.2% of the amount of DpC extracted from PBS by several independently prepared fibers. The SPME procedure was optimized by evaluating extraction and desorption time profiles. Suitability of the optimized protocol was verified by examining reproducibility, linearity, and recovery of DpC extracted from PBS or plasma. All samples extracted by SPME were analyzed using an optimized and validated UHPLC-MS/MS method. The developed procedure was applied to the in vitro determination of PPB of DpC at two clinically relevant concentrations (500 and 1000 ng/mL). These studies showed that DpC is highly bound to plasma proteins (PPB ≥ 88%) and this did not differ significantly between both concentrations tested. This investigation provides novel data in the applicability of SPME for the determination of PPB of chelators, as well as useful information for the clinical development of DpC. Graphical abstract.


Subject(s)
Antineoplastic Agents/metabolism , Blood Proteins/metabolism , Pyridines/metabolism , Solid Phase Microextraction/instrumentation , Thiosemicarbazones/metabolism , Adsorption , Animals , Cattle , Chromatography, High Pressure Liquid/methods , Equipment Design , Protein Binding , Rats , Silicon/chemistry , Solid Phase Microextraction/methods , Tandem Mass Spectrometry/methods
14.
Chem Res Toxicol ; 31(11): 1151-1163, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30395451

ABSTRACT

Salicylaldehyde isonicotinoyl hydrazone (SIH) is a small molecule and lipophilic chelating agent that firmly binds ferric ions from the cellular labile iron pool and is able to protect various tissues against oxidative damage. Previously, SIH possessed the best ratio of cytoprotective efficiency to toxicity among various iron chelators, including the desferrioxamine, deferiprone, and deferasirox used in clinical practice. Here, we prepared a series of 2,6-dihydroxybenzaldehyde aroylhydrazones as SIH analogues with an additional hydroxyl group that can be involved in the chelation of metal ions. Compound JK-31 (2,6-dihydroxybenzaldehyde 4-chlorobenzohydrazone) showed the best cytoprotective efficiency among the studied compounds including SIH. This compound significantly protected H9c2 cardiomyoblast cells against oxidative stress induced by various pro-oxidants, such as hydrogen peroxide, tert-butyl hydroperoxide, paraquat, epinephrine, N-acetyl- p-benzoquinone imine (a toxic metabolite of paracetamol), and 6-hydroxydopamine. The exceptional cytoprotective activity of JK-31 was confirmed using epifluorescence microscopy, where JK-31-treated H9c2 cells maintained a higher mitochondrial inner membrane potential in the presence of a lethal dose of hydrogen peroxide than was observed with cells treated with SIH. Hence, this study demonstrates the deleterious role of free iron ions in oxidative injury and the potential of 2,6-dihydroxybenzaldehyde aroylhydrazones in the prevention of various types of cardiac injuries, highlighting the need for further investigations into these compounds.


Subject(s)
Aldehydes/chemistry , Benzaldehydes/chemistry , Hydrazones/chemistry , Iron Chelating Agents/chemistry , Oxidative Stress , Aldehydes/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Stability , Humans , Hydrazones/pharmacology , Hydrolysis , Iron Chelating Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Protective Agents/chemistry , Protective Agents/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...