Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Article in English | MEDLINE | ID: mdl-35994895

ABSTRACT

Human leishmaniasis is an infectious disease caused by Leishmania protozoan parasites. Current chemotherapeutic options against the deadly disease have significant limitations. The ergosterol biosynthetic pathway has been identified as a drug target in Leishmania. However, remarkable differences in the efficacy of antifungal azoles that inhibit ergosterol biosynthesis have been reported for the treatment of leishmaniasis. To better understand the sterol biosynthetic pathway in Leishmania and elucidate the mechanism underlying the differential efficacy of antifungal azoles, we developed a new LC-MS/MS method to study sterol profiles in promastigotes of three Leishmania species, including two L. donovani, one L. major and one L. tarentolae strains. A combination of distinct precursor ion masses and LC retention times allowed for specific detection of sixteen intermediate sterols between lanosterol and ergosterol using the newly developed LC-MS/MS method. Although both posaconazole and fluconazole are known inhibitors of fungal lanosterol 14α-demethylase (CYP51), only posaconazole led to a substantial accumulation of lanosterol in azole-treated L. donovani promastigotes. Furthermore, a key intermediate sterol accumulated by 40- and 7-fold when these parasites were treated with posaconazole and fluconazole, respectively, which was determined as 4α,14α-dimethylzymosterol by high resolution mass spectrometry and NMR spectroscopy. The identification of 4α,14α-dimethylzymosterol supports a branched ergosterol biosynthetic pathway in Leishmania, where lanosterol C4- and C14-demethylation reactions occur in parallel rather than sequentially. Our results suggest that selective inhibition of leishmanial CYP51 is insufficient to effectively prevent parasite growth and dual inhibitors of both CYP51 and the unknown sterol C4-demethylase may be required for optimal antiparasitic effect.


Subject(s)
Leishmania , Parasites , Animals , Humans , Azoles/pharmacology , Ergosterol/pharmacology , Sterols/analysis , Sterols/pharmacology , Sterol 14-Demethylase , Biosynthetic Pathways , Tandem Mass Spectrometry , Parasites/metabolism , Chromatography, Liquid , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Lanosterol/analysis , Lanosterol/pharmacology
2.
Pharmacol Res Perspect ; 10(1): e00903, 2022 02.
Article in English | MEDLINE | ID: mdl-34918875

ABSTRACT

Metopimazine (MPZ) is a peripherally restricted, dopamine D2 receptor antagonist used for four decades to treat acute nausea and vomiting. MPZ is currently under clinical investigation for the treatment of gastroparesis (GP). MPZ undergoes high first-pass metabolism that produces metopimazine acid (MPZA), the major circulating metabolite in humans. Despite a long history of use, the enzymes involved in the metabolism of MPZ have not been identified. Here we report a series of studies designed to identify potential MPZ metabolites in vitro, determine their clinical relevance in humans, and elucidate the enzymes responsible for their formation. The findings demonstrated that the formation of MPZA was primarily catalyzed by human liver microsomal amidase. Additionally, human liver cytosolic aldehyde oxidase (AO) catalyzes the formation of MPZA, in vitro, although to a much lesser extent. Neither cytochrome P450 enzymes nor flavin-monooxygenases (FMO) were involved in the formation MPZA, although two minor oxidative pathways were catalyzed by CYP3A4 and CYP2D6 in vitro. Analysis of plasma samples from subjects dosed 60 mg of MPZ verified that these oxidative pathways are very minor and that CYP enzyme involvement was negligible compared to microsomal amidase/hydrolase in overall MPZ metabolism in humans. The metabolism by liver amidase, an enzyme family not well defined in small molecule drug metabolism, with minimal metabolism by CYPs, differentiates this drug from current D2 antagonists used or in development for the treatment of GP.


Subject(s)
Amidohydrolases/metabolism , Dopamine D2 Receptor Antagonists/metabolism , Isonipecotic Acids/metabolism , Microsomes, Liver/metabolism , Adolescent , Adult , Animals , Antiemetics/metabolism , Cohort Studies , Cytochrome P-450 Enzyme System/metabolism , Dogs , Double-Blind Method , Female , Humans , Male , Microsomes, Liver/enzymology , Middle Aged , Rats , Rats, Sprague-Dawley , Young Adult
3.
J Pharm Sci ; 108(12): 3870-3878, 2019 12.
Article in English | MEDLINE | ID: mdl-31545969

ABSTRACT

Eflornithine has been used to treat second-stage human African trypanosomiasis. However, it has inadequate oral bioavailability and low blood-brain barrier permeation, thus requiring a lengthy and complicated intravenous infusion schedule. Here, we investigated the feasibility of using an intercellular junction-modulating E-cadherin peptide HAV6 to enhance the oral bioavailability and blood-brain barrier permeation of eflornithine. Eflornithine was not metabolized in liver microsomes, nor was it a substrate for the human efflux transporter P-glycoprotein. Furthermore, HAV6 and HAV6scr (sequence scrambled HAV6) were stable in simulated gastric fluid with pepsin and rat intestinal mucosal scrapings. Both peptides were stable in human plasma, albeit less stable in rat and mouse plasma. HAV6 increased eflornithine permeability across Madin-Darby canine kidney and Caco-2 cell monolayers (5- and up to 8.5-fold), whereas HAV6scr had little effect. Using an in situ rat brain perfusion model, HAV6, but not HAV6scr, significantly increased eflornithine concentrations in different brain regions up to 4.9-fold. In rats, coadministration of HAV6 increased eflornithine oral bioavailability from 38% to 54%, brain concentrations by up to 83%, and cerebrospinal fluid concentrations by 40%. In conclusion, coadministration of HAV6, either during intravenous infusion or as an oral formulation, has the potential to improve eflornithine-based treatment for second-stage human African trypanosomiasis.


Subject(s)
Brain/metabolism , Cadherins/metabolism , Eflornithine/metabolism , Intercellular Junctions/metabolism , Peptides/metabolism , Administration, Oral , Animals , Biological Availability , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cell Line , Cell Line, Tumor , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Permeability , Rats , Rats, Sprague-Dawley
4.
Article in English | MEDLINE | ID: mdl-29061761

ABSTRACT

Given the limitations of current antileishmanial drugs and the utility of oral combination therapy for other infections, developing an oral combination against visceral leishmaniasis should be a high priority. In vitro combination studies with DB766 and antifungal azoles against intracellular Leishmania donovani showed that posaconazole and ketoconazole, but not fluconazole, enhanced DB766 potency. Pharmacokinetic analysis of DB766-azole combinations in uninfected Swiss Webster mice revealed that DB766 exposure was increased by higher posaconazole and ketoconazole doses, while DB766 decreased ketoconazole exposure. In L. donovani-infected BALB/c mice, DB766-posaconazole combinations given orally for 5 days were more effective than DB766 or posaconazole alone. For example, 81% ± 1% (means ± standard errors) inhibition of liver parasite burden was observed for 37.5 mg/kg of body weight DB766 plus 15 mg/kg posaconazole, while 37.5 mg/kg DB766 and 15 mg/kg posaconazole administered as monotherapy gave 40% ± 5% and 21% ± 3% inhibition, respectively. Combination index (CI) analysis indicated that synergy or moderate synergy was observed in six of nine combined dose groups, while the other three were nearly additive. Liver concentrations of DB766 and posaconazole increased in almost all combination groups compared to monotherapy groups, although many increases were not statistically significant. For DB766-ketoconazole combinations evaluated in this model, two were antagonistic, one displayed synergy, and one was nearly additive. These data indicate that the efficacy of DB766-posaconazole and DB766-ketoconazole combinations in vivo is influenced in part by the pharmacokinetics of the combination, and that the former combination deserves further consideration in developing new treatment strategies against visceral leishmaniasis.


Subject(s)
Amidines/pharmacology , Antiprotozoal Agents/pharmacology , Furans/pharmacology , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Amidines/pharmacokinetics , Animals , Antiprotozoal Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Synergism , Drug Therapy, Combination , Female , Furans/pharmacokinetics , Ketoconazole/pharmacokinetics , Ketoconazole/pharmacology , Leishmania donovani/metabolism , Mice , Mice, Inbred BALB C , Protozoan Proteins/metabolism , Triazoles/pharmacokinetics , Triazoles/pharmacology
5.
Br J Pharmacol ; 174(15): 2576-2590, 2017 08.
Article in English | MEDLINE | ID: mdl-28542844

ABSTRACT

BACKGROUND AND PURPOSE: The density of the inward rectifier current (IK1 ) increases in atrial fibrillation (AF), shortening effective refractory period and thus promoting atrial re-entry. The synthetic compound pentamidine analogue 6 (PA-6) is a selective and potent IK1 inhibitor. We tested PA-6 for anti-AF efficacy and potential proarrhythmia, using established models in large animals. EXPERIMENTAL APPROACH: PA-6 was applied i.v. in anaesthetized goats with rapid pacing-induced AF and anaesthetized dogs with chronic atrio-ventricular (AV) block. Electrophysiological and pharmacological parameters were determined. KEY RESULTS: PA-6 (2.5 mg·kg-1 ·10 min-1 ) induced cardioversion to sinus rhythm (SR) in 5/6 goats and prolonged AF cycle length. AF complexity decreased significantly before cardioversion. PA-6 accumulated in cardiac tissue with ratios between skeletal muscle : atrial muscle : ventricular muscle of approximately 1:8:21. In SR dogs, PA-6 peak plasma levels 10 min post infusion were 5.5 ± 0.9 µM, PA-6 did not induce significant prolongation of QTc and did not affect heart rate, PQ or QRS duration. In dogs with chronic AV block, PA-6 did not affect QRS but lengthened QTc during the experiment, but not chronically. PA-6 did not induce TdP arrhythmias in nine animals (0/9) in contrast to dofetilide (5/9). PA-6 (200 nM) inhibited IK1 , but not IK,ACh , in human isolated atrial cardiomyocytes. CONCLUSION AND IMPLICATIONS: PA-6 restored SR in goats with persistent AF and, in dogs with chronic AV block, prolonged QT intervals, without inducing TdP arrhythmias. Our results demonstrate cardiac safety and good anti-AF properties for PA-6.


Subject(s)
Atrial Fibrillation/chemically induced , Disease Models, Animal , Pentamidine/pharmacology , Administration, Intravenous , Animals , Dogs , Female , Goals , Humans , Pentamidine/administration & dosage , Pentamidine/analogs & derivatives
6.
Bioorg Med Chem Lett ; 26(10): 2551-2556, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27048943

ABSTRACT

Arylimidamide (AIA) compounds containing two pyridylimidamide terminal groups (bis-AIAs) possess outstanding in vitro antileishmanial activity, and the frontrunner bis-AIA DB766 (2,5-bis[2-(2-isopropoxy)-4-(2-pyridylimino)aminophenyl]furan) is active in visceral leishmaniasis models when given orally. Eighteen compounds containing a single pyridylimidamide terminal group (mono-AIAs) were synthesized and evaluated for their antileishmanial potential. Six of these compounds exhibited sub-micromolar potency against both intracellular Leishmania donovani and Leishmania amazonensis amastigotes, and three of these compounds also displayed selectivity indexes of 25 or greater for the parasites compared to a J774 macrophage cell line. When given orally at a dose of 100mg/kg/day for five days, compound 1b (N-(3-isopropoxy-4-(5-phenylfuran-2-yl)phenyl)picolinimidamide methanesulfonate) reduced liver parasitemia by 46% in L. donovani-infected mice. Mono-AIAs are thus a new class of candidate molecules for antileishmanial drug development.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Leishmania donovani/drug effects , Leishmania mexicana/drug effects , Administration, Oral , Animals , Antiprotozoal Agents/chemical synthesis , Chemistry Techniques, Synthetic , Drug Evaluation, Preclinical/methods , Furans/chemistry , Inhibitory Concentration 50 , Leishmania donovani/pathogenicity , Leishmania mexicana/pathogenicity , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/parasitology , Macrophages/drug effects , Macrophages/parasitology , Mice, Inbred BALB C , Parasitemia/drug therapy , Parasitemia/parasitology , Structure-Activity Relationship
7.
Bioorg Med Chem ; 24(11): 2451-65, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27102161

ABSTRACT

2-(2-Benzamido)ethyl-4-phenylthiazole (1) was one of 1035 molecules (grouped into 115 distinct scaffolds) found to be inhibitory to Trypanosoma brucei, the pathogen causing human African trypanosomiasis, at concentrations below 3.6µM and non-toxic to mammalian (Huh7) cells in a phenotypic high-throughput screen of a 700,000 compound library performed by the Genomics Institute of the Novartis Research Foundation (GNF). Compound 1 and 72 analogues were synthesized in this lab by one of two general pathways. These plus 10 commercially available analogues were tested against T. brucei rhodesiense STIB900 and L6 rat myoblast cells (for cytotoxicity) in vitro. Forty-four derivatives were more potent than 1, including eight with IC50 values below 100nM. The most potent and most selective for the parasite was the urea analogue 2-(2-piperidin-1-ylamido)ethyl-4-(3-fluorophenyl)thiazole (70, IC50=9nM, SI>18,000). None of 33 compounds tested were able to cure mice infected with the parasite; however, seven compounds caused temporary reductions of parasitemia (⩾97%) but with subsequent relapses. The lack of in vivo efficacy was at least partially due to their poor metabolic stability, as demonstrated by the short half-lives of 15 analogues against mouse and human liver microsomes.


Subject(s)
Amides/pharmacology , Amines/pharmacology , Antiprotozoal Agents/pharmacology , Thiazoles/pharmacology , Trypanosoma brucei rhodesiense/drug effects , Urea/pharmacology , Amides/chemistry , Amines/chemical synthesis , Amines/chemistry , Animals , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Myoblasts/drug effects , Parasitic Sensitivity Tests , Rats , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Urea/analogs & derivatives , Urea/chemistry
9.
Bioorg Med Chem ; 23(16): 5182-9, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25749014

ABSTRACT

Visceral leishmaniasis is a neglected parasitic disease that has a high fatality rate in the absence of treatment. New drugs that are inexpensive, orally active, and effective could be useful tools in the fight against this disease. We previously showed that N(2),N(4)-disubstituted quinazoline-2,4-diamines displayed low- to sub-micromolar potency against intracellular Leishmania, and lead compound N(4)-(furan-2-ylmethyl)-N(2)-isopropyl-7-methylquinazoline-2,4-diamine (4) exhibited modest efficacy in an acute murine model of visceral leishmaniasis. In the present work, thirty-one N(2),N(4)-disubstituted quinazoline-2,4-diamines that had not previously been examined for their antileishmanial activity were evaluated for their potency and selectivity against Leishmania donovani, the causative parasite of visceral leishmaniasis. Quinazoline-2,4-diamines with aromatic substituents at both N(2) and N(4) exhibited potent in vitro antileishmanial activity but relatively low selectivity, while compounds substituted with small alkyl groups at either N(2) or N(4) generally showed lower antileishmanial potency but were less toxic to a murine macrophage cell line. Based on their in vitro antileishmanial potency, N(4)-benzyl-N(2)-(4-chlorobenzyl)quinazoline-2,4-diamine (15) and N(2)-benzyl-N(4)-isopropylquinazoline-2,4-diamine (40) were selected for in vivo evaluation of their pharmacokinetic and antileishmanial properties. While 15 displayed a longer plasma half-life and a greater area under the curve than 40, both compounds showed low efficacy in an acute murine visceral leishmaniasis model. Although the present study did not identify new quinazoline-2,4-diamines with promising in vivo efficacy, the reduced in vitro toxicity of derivatives bearing small alkyl groups at either N(2) or N(4) may provide clues for the design of safe and effective antileishmanial quinazolines.


Subject(s)
Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/therapeutic use , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Neglected Diseases/drug therapy , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Diamines/chemistry , Diamines/pharmacokinetics , Diamines/pharmacology , Diamines/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship
10.
Biomed Chromatogr ; 29(6): 918-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25365958

ABSTRACT

Eflornithine (α-difluoromethylornithine) has been used to treat second-stage (or meningoencephalitic-stage) human African trypanosomiasis and currently is under clinical development for cancer prevention. In this study, a new ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS)-based assay was developed and validated for the quantification of eflornithine in rat brain. To improve chromatographic retention and MS detection, eflornithine was derivatized with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate for 5 min at room temperature prior to injection. Derivatized eflornithine was separated on a reverse-phase C18 UPLC column with a 6-min gradient; elution occurred at approximately 1.5 min. Prior to derivatization, eflornithine was reproducibly extracted from rat brain homogenate by methanol protein precipitation (~70% recovery). Derivatized eflornithine was stable in the autosampler (6 °C) for at least 24 h. This new assay had acceptable intra- and interday accuracy and precision over a wide dynamic range (5000-fold) and excellent sensitivity with a lower limit of quantification of 0.1 µm (18 ng/mL) using only 10 µL of rat brain homogenate. The validated eflornithine assay was applied successfully to determine eflornithine distribution in different regions of rat brain in an in situ rat brain perfusion study.


Subject(s)
Brain/metabolism , Chromatography, High Pressure Liquid/methods , Eflornithine/analysis , Tandem Mass Spectrometry/methods , Trypanocidal Agents/analysis , Animals , Brain Chemistry , Eflornithine/chemistry , Eflornithine/pharmacokinetics , Linear Models , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacokinetics
11.
Antimicrob Agents Chemother ; 58(7): 4064-74, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798280

ABSTRACT

Human African trypanosomiasis (HAT), a neglected tropical disease, is fatal without treatment. Pentamidine, a cationic diamidine, has been used to treat first-stage (hemolymphatic) HAT since the 1940s, but it is ineffective against second-stage (meningoencephalitic, or central nervous system [CNS]) infection. Novel diamidines (DB75, DB820, and DB829) have shown promising efficacy in both mouse and monkey models of first-stage HAT. However, only DB829 cured animals with second-stage infection. In this study, we aimed to determine the mechanisms underlying the differential efficacies of these diamidines against HAT by conducting a comprehensive pharmacokinetic characterization. This included the determination of metabolic stability in liver microsomes, permeability across MDCK and MDR1-MDCK cell monolayers, interaction with the efflux transporter MDR1 (P-glycoprotein 1 or P-gp), drug binding in plasma and brain, and plasma and brain concentration-time profiles after a single dose in mice. The results showed that DB829, an azadiamidine, had the highest systemic exposure and brain-to-plasma ratio, whereas pentamidine and DB75 had the lowest. None of these diamidines was a P-gp substrate, and the binding of each to plasma proteins and brain differed greatly. The brain-to-plasma ratio best predicted the relative efficacies of these diamidines in mice with second-stage infection. In conclusion, pharmacokinetics and CNS penetration influenced the in vivo efficacies of cationic diamidines against first- and second-stage HAT and should be considered when developing CNS-active antitrypanosomal diamidines.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amidines/pharmacokinetics , Pentamidine/pharmacokinetics , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei rhodesiense/drug effects , Amidines/metabolism , Amidines/pharmacology , Animals , Benzamidines/metabolism , Benzamidines/pharmacokinetics , Benzamidines/pharmacology , Blood-Brain Barrier , Cell Line , Dogs , Furans/metabolism , Furans/pharmacokinetics , Furans/pharmacology , Madin Darby Canine Kidney Cells , Male , Mice , Pentamidine/metabolism , Pentamidine/pharmacology , Protein Binding , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
12.
J Med Chem ; 57(12): 5141-56, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24874647

ABSTRACT

A series of N(2),N(4)-disubstituted quinazoline-2,4-diamines has been synthesized and tested against Leishmania donovani and L. amazonensis intracellular amastigotes. A structure-activity and structure-property relationship study was conducted in part using the Topliss operational scheme to identify new lead compounds. This study led to the identification of quinazolines with EC50 values in the single digit micromolar or high nanomolar range in addition to favorable physicochemical properties. Quinazoline 23 also displayed efficacy in a murine model of visceral leishmaniasis, reducing liver parasitemia by 37% when given by the intraperitoneal route at 15 mg kg(-1) day(-1) for 5 consecutive days. Their antileishmanial efficacy, ease of synthesis, and favorable physicochemical properties make the N(2),N(4)-disubstituted quinazoline-2,4-diamine compound series a suitable platform for future development of antileishmanial agents.


Subject(s)
Diamines/chemistry , Leishmania/drug effects , Quinazolines/chemistry , Trypanocidal Agents/chemistry , Animals , Antimony/pharmacology , Cell Line , Diamines/pharmacokinetics , Diamines/pharmacology , Drug Resistance , Leishmania donovani/drug effects , Leishmania donovani/isolation & purification , Leishmania mexicana/drug effects , Leishmaniasis, Visceral/drug therapy , Macrophages/drug effects , Macrophages/parasitology , Male , Mice, Inbred BALB C , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Structure-Activity Relationship , Trypanocidal Agents/pharmacokinetics , Trypanocidal Agents/pharmacology
13.
Antimicrob Agents Chemother ; 58(8): 4452-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24867978

ABSTRACT

African sleeping sickness is a neglected tropical disease transmitted by tsetse flies. New and better drugs are still needed especially for its second stage, which is fatal if untreated. 28DAP010, a dipyridylbenzene analogue of DB829, is the second simple diamidine found to cure mice with central nervous system infections by a parenteral route of administration. 28DAP010 showed efficacy similar to that of DB829 in dose-response studies in mouse models of first- and second-stage African sleeping sickness. The in vitro time to kill, determined by microcalorimetry, and the parasite clearance time in mice were shorter for 28DAP010 than for DB829. No cross-resistance was observed between 28DAP010 and pentamidine on the tested Trypanosoma brucei gambiense isolates from melarsoprol-refractory patients. 28DAP010 is the second promising preclinical candidate among the diamidines for the treatment of second-stage African sleeping sickness.


Subject(s)
Amidines/pharmacology , Pyridines/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei gambiense/drug effects , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy , Amidines/chemical synthesis , Amidines/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Female , Humans , Melarsoprol/pharmacokinetics , Melarsoprol/pharmacology , Mice , Pentamidine/pharmacokinetics , Pentamidine/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei gambiense/growth & development , Trypanosoma brucei gambiense/pathogenicity , Trypanosoma brucei rhodesiense/growth & development , Trypanosoma brucei rhodesiense/pathogenicity , Trypanosomiasis, African/parasitology
14.
J Pharm Sci ; 103(1): 337-49, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24186380

ABSTRACT

DB844 (CPD-594-12), N-methoxy-6-{5-[4-(N-methoxyamidino)phenyl]-furan-2-yl}-nicotinamidine, is an oral prodrug that has shown promising efficacy in both mouse and monkey models of second stage human African trypanosomiasis. However, gastrointestinal (GI) toxicity was observed with high doses in a vervet monkey safety study. In the current study, we compared the metabolism of DB844 by hepatic and extrahepatic cytochrome P450s to determine whether differences in metabolite formation underlie the observed GI toxicity. DB844 undergoes sequential O-demethylation and N-dehydroxylation in the liver to form the active compound DB820 (CPD-593-12). However, extrahepatic CYP1A1 and CYP1B1 produced two new metabolites, MX and MY. Accurate mass and collision-induced dissociation mass spectrometry analyses of the metabolites supported proposed structures of MX and MY. In addition, MY was confirmed with a synthetic standard and detection of nitric oxide (NO) release when DB844 was incubated with CYP1A1. Taken altogether, we propose that MX is formed by insertion of oxygen into the amidine CN to form an oxaziridine, which is followed by intramolecular rearrangement of the adjacent O-methyl group and subsequent release of NO. The resulting imine ester, MX, is further hydrolyzed to form MY. These findings may contribute to furthering the understanding of toxicities associated with benzamidoxime- and benzmethamidoxime-containing molecules.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Benzamidines/metabolism , Biotransformation/physiology , Cytochrome P-450 CYP1A1/metabolism , Furans/metabolism , Prodrugs/metabolism , Animals , Cytochrome P-450 CYP1B1 , Haplorhini/metabolism , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Microsomes, Liver
15.
Xenobiotica ; 44(1): 77-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23808362

ABSTRACT

1. Spontaneously hypertensive rats (SHRs) and deoxycorticosterone acetate-salt-induced hypertensive rats (DOCA-salt rats) have been developed as animal models for human essential (idiopathic or primary) and secondary hypertensions, respectively. 2. In order to identify pharmacokinetic changes (mainly non-renal clearance, CLNR) in 16-week-old SHRs due to hereditary characteristics and/or neither the hypertensive state itself, we reviewed the pharmacokinetics of drugs in 6- (blood pressure within a normotensive range) and 16-week-old SHRs and 16-week-old DOCA-salt rats compared with respective control rats. 3. We reviewed changes in CLNRs of drugs which are primarily metabolized via hepatic microsomal cytochrome P 450 enzymes (CYPs) based mainly on data from hypertensive rats, and present the data in terms of changes in in vitro hepatic intrinsic clearance (CLint), free fraction in plasma (fp) and hepatic blood flow rate (QH) depending on the hepatic excretion ratios of drugs. In general, changes in the CLNRs of drugs in this category were well-explained by the above-described factors. 4. We also reviewed and discussed the mechanism of urinary excretion of drugs (i.e. glomerular filtration and active renal secretion or reabsorption) in hypertensive rats.


Subject(s)
Desoxycorticosterone Acetate/adverse effects , Disease Models, Animal , Hypertension/chemically induced , Hypertension/metabolism , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Rats, Inbred SHR/metabolism , Age Factors , Animals , Cytochrome P-450 Enzyme System/metabolism , Glomerular Filtration Rate , Metabolic Clearance Rate , Models, Biological , Rats
16.
Bioorg Med Chem ; 21(21): 6732-41, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24012380

ABSTRACT

Dicationic 2,6-diphenylpyrazines, aza-analogues and prodrugs were synthesized; evaluated for DNA affinity, activity against Trypanosoma brucei rhodesiense (T. b. r.) and Plasmodium falciparum (P. f.) in vitro, efficacy in T. b. r. STIB900 acute and T. b. brucei GVR35 CNS mouse models. Most diamidines gave poly(dA-dT)2 ΔTm values greater than pentamidine, IC50 values: T. b. r. (4.8-37nM) and P. f. (10-52nM). Most diamidines and prodrugs gave cures for STIB900 model (11, 19a and 24b 4/4 cures); 12 3/4 cures for GVR35 model. Metabolic stability half-life values for O-methylamidoxime prodrugs did not correlate with STIB900 results.


Subject(s)
Antiprotozoal Agents/chemical synthesis , Aza Compounds/chemistry , Pentamidine/chemistry , Prodrugs/chemical synthesis , Pyrazines/chemistry , Animals , Antiprotozoal Agents/therapeutic use , Antiprotozoal Agents/toxicity , Cations/chemistry , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Mice , Myoblasts/cytology , Parasitic Sensitivity Tests , Pentamidine/therapeutic use , Pentamidine/toxicity , Plasmodium falciparum/drug effects , Poly dA-dT/chemistry , Poly dA-dT/metabolism , Prodrugs/therapeutic use , Prodrugs/toxicity , Rats , Structure-Activity Relationship , Transition Temperature , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy
17.
Antimicrob Agents Chemother ; 57(11): 5330-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959303

ABSTRACT

Human African trypanosomiasis (HAT, also called sleeping sickness), a neglected tropical disease endemic to sub-Saharan Africa, is caused by the parasites Trypanosoma brucei gambiense and T. brucei rhodesiense. Current drugs against this disease have significant limitations, including toxicity, increasing resistance, and/or a complicated parenteral treatment regimen. DB829 is a novel aza-diamidine that demonstrated excellent efficacy in mice infected with T. b. rhodesiense or T. b. brucei parasites. The current study examined the pharmacokinetics, in vitro and in vivo activity against T. b. gambiense, and time of drug action of DB829 in comparison to pentamidine. DB829 showed outstanding in vivo efficacy in mice infected with parasites of T. b. gambiense strains, despite having higher in vitro 50% inhibitory concentrations (IC50s) than against T. b. rhodesiense strain STIB900. A single dose of DB829 administered intraperitoneally (5 mg/kg of body weight) cured all mice infected with different T. b. gambiense strains. No cross-resistance was observed between DB829 and pentamidine in T. b. gambiense strains isolated from melarsoprol-refractory patients. Compared to pentamidine, DB829 showed a greater systemic exposure when administered intraperitoneally, partially contributing to its improved efficacy. Isothermal microcalorimetry and in vivo time-to-kill studies revealed that DB829 is a slower-acting trypanocidal compound than pentamidine. A single dose of DB829 (20 mg/kg) administered intraperitoneally clears parasites from mouse blood within 2 to 5 days. In summary, DB829 is a promising preclinical candidate for the treatment of first- and second-stage HAT caused by both Trypanosoma brucei subspecies.


Subject(s)
Amidines/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei gambiense/drug effects , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy , Amidines/pharmacokinetics , Animals , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Humans , Injections, Intraperitoneal , Melarsoprol/pharmacology , Mice , Parasitic Sensitivity Tests , Pentamidine/pharmacology , Time Factors , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei gambiense/growth & development , Trypanosoma brucei rhodesiense/growth & development , Trypanosomiasis, African/parasitology
18.
Antimicrob Agents Chemother ; 56(7): 3690-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22508306

ABSTRACT

Arylimidamides (AIAs) have shown outstanding in vitro potency against intracellular kinetoplastid parasites, and the AIA 2,5-bis[2-(2-propoxy)-4-(2-pyridylimino)aminophenyl]furan dihydrochloride (DB766) displayed good in vivo efficacy in rodent models of visceral leishmaniasis (VL) and Chagas' disease. In an attempt to further increase the solubility and in vivo antikinetoplastid potential of DB766, the mesylate salt of this compound and that of the closely related AIA 2,5-bis[2-(2-cyclopentyloxy)-4-(2-pyridylimino)aminophenyl]furan hydrochloride (DB1852) were prepared. These two mesylate salts, designated DB1960 and DB1955, respectively, exhibited dose-dependent activity in the murine model of VL, with DB1960 inhibiting liver parasitemia by 51% at an oral dose of 100 mg/kg/day × 5 and DB1955 reducing liver parasitemia by 57% when given by the same dosing regimen. In a murine Trypanosoma cruzi infection model, DB1960 decreased the peak parasitemia levels that occurred at 8 days postinfection by 46% when given orally at 100 mg/kg/day × 5, while DB1955 had no effect on peak parasitemia levels when administered by the same dosing regimen. Distribution studies revealed that these compounds accumulated to micromolar levels in the liver, spleen, and kidneys but to a lesser extent in the heart, brain, and plasma. A 5-day repeat-dose toxicology study with DB1960 and DB1955 was also conducted with female BALB/c mice, with the compounds administered orally at 100, 200, and 500 mg/kg/day. In the high-dose groups, DB1960 caused changes in serum chemistry, with statistically significant increases in serum blood urea nitrogen, lactate dehydrogenase, aspartate aminotransferase, and alanine aminotransferase levels, and a 21% decrease in body weight was observed in this group. These changes were consistent with microscopic findings in the livers and kidneys of the treated animals. The incidences of observed clinical signs (hunched posture, tachypnea, tremors, and ruffled fur) were more frequent in DB1960-treated groups than in those treated with DB1955. However, histopathological examination of tissue samples indicated that both compounds had adverse effects at all dose levels.


Subject(s)
Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/therapeutic use , Chagas Disease/drug therapy , Leishmaniasis, Visceral/drug therapy , Amidines/therapeutic use , Animals , Antiprotozoal Agents/pharmacology , Disease Models, Animal , Female , Furans/therapeutic use , Male , Mice , Mice, Inbred BALB C , Parasitemia/drug therapy , Solubility , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/pathogenicity
19.
Acta Pharmacol Sin ; 32(7): 967-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21666702

ABSTRACT

AIM: Losartan and antiplatelet agent ticlopidine can be prescribed concomitantly for prevention or therapy of cardiovascular diseases. Hence, the effects of ticlopidine on the pharmacokinetics of losartan and its active metabolite EXP-3174 were evaluated in rats. METHODS: Ticlopidine (4 or 10 mg/kg po) was administered 30 min before administration of losartan (9 mg/kg po or 3 mg/kg iv). The activity of human CYP2C9 and 3A4 were measured using the CYP inhibition assay kit. The activity of P-gp was evaluated using rhodamine-123 retention assay in MCF-7/ADR cells. RESULTS: Ticlopidine (10 mg/kg) significantly increased the areas under the plasma concentration-time curves (AUCs) and peak plasma concentration (C(max)) of oral losartan (9 mg/kg), as well as the AUCs of the active metabolite EXP-3174. Ticlopidine (10 mg/kg) did not significantly change the pharmacokinetics of intravenous losartan (3 mg/kg). Ticlopidine inhibited CYP2C9 and 3A4 with IC50 values of 26.0 and 32.3 µmol/L, respectively. The relative cellular uptake of rhodamine-123 was unchanged. CONCLUSION: The significant increase in the AUC of losartan (9 mg/kg) by ticlopidine (10 mg/kg) could be attributed to the inhibition of CYP2C9- and 3A4-mediated losartan metabolism in small intestine and/or in liver. The inhibition of P-gp in small intestine and reduction of renal elimination of losartan by ticlopidine are unlikely to be causal factors.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Losartan/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Tetrazoles/pharmacokinetics , Ticlopidine/pharmacology , Animals , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Line , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Drug Interactions , Humans , Inhibitory Concentration 50 , Male , Rats , Rats, Sprague-Dawley
20.
Pharmacology ; 88(1-2): 1-9, 2011.
Article in English | MEDLINE | ID: mdl-21709429

ABSTRACT

The present study was designed to investigate the effects of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (atorvastatin, pravastatin, simvastatin) on the pharmacokinetics of losartan and its active metabolite EXP-3174 in rats. Pharmacokinetic parameters of losartan and EXP-3174 in rats were determined after oral and intravenous administration of losartan (9 mg/kg) without and with HMG-CoA reductase inhibitors (1 mg/kg). The effect of HMG-CoA reductase inhibitors on P-gp and cytochrome (CYP) 3A4 activity were also evaluated. Atorvastatin, pravastatin and simvastatin inhibited CYP3A4 activities with IC50 values of 48.0, 14.1 and 3.10 µmol/l, respectively. Simvastatin (1-10 µmol/l) enhanced the cellular uptake of rhodamine-123 in a concentration-dependent manner. The area under the plasma concentration-time curve (AUC0₋∞) and the peak plasma concentration of losartan were significantly (p < 0.05) increased by 59.6 and 45.8%, respectively, by simvastatin compared to those of control. The total body clearance (CL/F) of losartan after oral administration with simvastatin was significantly decreased (by 34.8%) compared to that of controls. Consequently, the absolute bioavailability (F) of losartan after oral administration with simvastatin was significantly increased by 59.4% compared to that of control. The metabolite-parent AUC ratio was significantly decreased by 25.7%, suggesting that metabolism of losartan was inhibited by simvastatin. In conclusion, the enhanced bioavailability of losartan might be mainly due to inhibition of P-gp in the small intestine and CYP3A subfamily-mediated metabolism of losartan in the small intestine and/or liver and to reduction of the CL/F of losartan by simvastatin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Anticholesteremic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacokinetics , Losartan/pharmacokinetics , Tetrazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acyl Coenzyme A/antagonists & inhibitors , Administration, Oral , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacology , Cell Line, Tumor , Cytochrome P-450 CYP3A Inhibitors , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Imidazoles/blood , Imidazoles/pharmacology , Injections, Intravenous , Losartan/administration & dosage , Losartan/metabolism , Losartan/pharmacology , Lovastatin/analogs & derivatives , Lovastatin/pharmacokinetics , Lovastatin/pharmacology , Male , Rats , Rats, Sprague-Dawley , Rhodamine 123/metabolism , Simvastatin/blood , Simvastatin/metabolism , Simvastatin/pharmacology , Tetrazoles/blood , Tetrazoles/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...