Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Biosci Biotechnol Biochem ; 88(6): 648-655, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38490741

ABSTRACT

Lysophosphatidylcholine (LPC) is present in various foods and contains a choline moiety such as in glycerophosphocholine (GPC). However, the potential of LPC as a choline source remains unclear. This study investigated the single-dose pharmacokinetics of 480 mg soy-derived LPC in 12 healthy men compared with that of either soy oil with the same lipid amount (placebo) or GPC with the same choline amount. Both LPC and GPC supplementation increased plasma choline, serum phospholipid, and serum triglyceride concentrations, but neither of them significantly elevated plasma trimethylamine N-oxide concentration. In addition, although the intake of LPC slightly increased plasma LPC16:0, LPC18:2, and total LPC concentrations, their concentrations remained within physiological ranges. No adverse events were attributed to the LPC supplementation. To the best of our knowledge, this study is the first to compare LPC and GPC pharmacokinetics in humans and shows that LPC can be a source of choline.


Subject(s)
Choline , Glycerylphosphorylcholine , Glycine max , Lysophosphatidylcholines , Humans , Male , Lysophosphatidylcholines/blood , Glycerylphosphorylcholine/pharmacokinetics , Glycerylphosphorylcholine/blood , Choline/pharmacokinetics , Choline/blood , Adult , Glycine max/chemistry , Dietary Supplements , Young Adult , Triglycerides/blood , Methylamines/blood , Methylamines/pharmacokinetics
2.
Drug Metab Dispos ; 49(12): 1109-1117, 2021 12.
Article in English | MEDLINE | ID: mdl-34625435

ABSTRACT

Linerixibat, an oral small-molecule ileal bile acid transporter inhibitor under development for cholestatic pruritus in primary biliary cholangitis, was designed for minimal absorption from the intestine (site of pharmacological action). This study characterized the pharmacokinetics, absorption, metabolism, and excretion of [14C]-linerixibat in humans after an intravenous microtracer concomitant with unlabeled oral tablets and [14C]-linerixibat oral solution. Linerixibat exhibited absorption-limited flip-flop kinetics: longer oral versus intravenous half-life (6-7 hours vs. 0.8 hours). The short intravenous half-life was consistent with high systemic clearance (61.9 l/h) and low volume of distribution (16.3 l). In vitro studies predicted rapid hepatic clearance via cytochrome P450 3A4 metabolism, which predicted human hepatic clearance within 1.5-fold. However, linerixibat was minimally metabolized in humans after intravenous administration: ∼80% elimination via biliary/fecal excretion (>90%-97% as unchanged parent) and ∼20% renal elimination by glomerular filtration (>97% as unchanged parent). Absolute oral bioavailability of linerixibat was exceedingly low (0.05%), primarily because of a very low fraction absorbed (0.167%; fraction escaping first-pass gut metabolism (fg) ∼100%), with high hepatic extraction ratio (77.0%) acting as a secondary barrier to systemic exposure. Oral linerixibat was almost entirely excreted (>99% recovered radioactivity) in feces as unchanged and unabsorbed linerixibat. Consistent with the low oral fraction absorbed and ∼20% renal recovery of intravenous [14C]-linerixibat, urinary elimination of orally administered radioactivity was negligible (<0.04% of dose). Linerixibat unequivocally exhibited minimal gastrointestinal absorption and oral systemic exposure. Linerixibat represents a unique example of high CYP3A4 clearance in vitro but nearly complete excretion as unchanged parent drug via the biliary/fecal route. SIGNIFICANCE STATEMENT: This study conclusively established minimal absorption and systemic exposure to orally administered linerixibat in humans. The small amount of linerixibat absorbed was eliminated efficiently as unchanged parent drug via the biliary/fecal route. The hepatic clearance mechanism was mispredicted to be mediated via cytochrome P450 3A4 metabolism in vitro rather than biliary excretion of unchanged linerixibat in vivo.


Subject(s)
Administration, Intravenous , Administration, Oral , Carrier Proteins/antagonists & inhibitors , Hepatobiliary Elimination , Membrane Glycoproteins/antagonists & inhibitors , Methylamines/pharmacokinetics , Renal Elimination , Thiazepines/pharmacokinetics , Adult , Biological Availability , Gastrointestinal Agents/pharmacokinetics , Healthy Volunteers , Hepatobiliary Elimination/drug effects , Hepatobiliary Elimination/physiology , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Renal Elimination/drug effects , Renal Elimination/physiology , Treatment Outcome
3.
Eur J Pharmacol ; 909: 174362, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34297968

ABSTRACT

The functions and mechanisms of GPR40 receptor to ameliorating the Alzheimer's disease (AD) by external treatment of encephalopathy remain unknown. In present study, the typical Aß1-42 induced mice model was applied to explore the functions and mechanisms of GPR40 receptor by external treatment of encephalopathy in AD. GPR40 agonist GW9508 and antagonist GW1100 were given by i.g injection to activate/inhibit the GPR40 receptor respectively in the gut of AD mouse which illustrated the function and mechanism of GPR40 receptor in ameliorating AD symptoms by external treatment of encephalopathy. A series of behavioral experiments were used to investigate the cognitive function and memory ability of mice, while molecular biology experiments such as Western blot, ELISA, flow cytometry were used to detect the corresponding changes of signaling pathways. The results revealed that intragastric administrated GW9508 could significantly ameliorate cognitive deficits of AD mouse, up-regulate the expression levels of gut-brain peptides both in blood circulation and hypothalamus thus up-regulate the expression levels of α-MSH in hypothalamus, while the negative autophagy-related proteins and inflammation-related proteins were down-regulated correspondingly. Meanwhile, GW9508 could also inhibit the pathological process of neuroinflammation in microglia. GW1100 reversed the effects of GW9508 significantly. These results suggested that GPR40 was an underlying therapeutic target for the external treatment of encephalopathy related to AD and GPR40 agonist could be explored as the emerging AD therapeutic drug.


Subject(s)
Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Methylamines/administration & dosage , Neuroinflammatory Diseases/drug therapy , Propionates/administration & dosage , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Alzheimer Disease/complications , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/toxicity , Animals , Behavior Observation Techniques , Blood-Brain Barrier/metabolism , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Down-Regulation/drug effects , Humans , Hypothalamus/drug effects , Hypothalamus/immunology , Hypothalamus/pathology , Male , Methylamines/pharmacokinetics , Mice , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/diagnosis , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Propionates/pharmacokinetics , Receptors, G-Protein-Coupled/metabolism , Tissue Distribution
4.
Drug Metab Pharmacokinet ; 37: 100369, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33513464

ABSTRACT

Flavin-containing monooxygenase (FMO) catalyzes the oxygenation of a wide variety of medicines and dietary-derived compounds. However, little information is available regarding drug interactions mediated by FMO3 in vivo. Consequently, we investigated interactions between FMO substrates in humanized-liver mice. Trimethylamine-d9 and itopride were, respectively, intravenously and orally administered to humanized-liver mice (n = 5-7). The pharmacokinetic profiles of itopride (the victim drug) in the presence of trimethylamine (the perpetrator drug) were determined for 24 h after co-administration using liquid chromatography/tandem mass spectrometry. Itopride (10 mg/kg) was extensively oxygenated in humanized-liver mice to its N-oxide. The plasma concentrations of itopride N-oxide after co-administration of itopride and trimethylamine (10 and 100 mg/kg) were significantly suppressed in a dose-dependent manner, but only during the early phase, i.e., up to 2 h after co-administration. With the higher dose of trimethylamine, the areas under the concentration-time curves of itopride and its N-oxide significantly increased (1.6-fold) and decreased (to 60%), respectively; modeling suggested that these modified pharmacokinetics resulted from suppression of the in vivo hepatic intrinsic clearance (to 67%). These results suggest that food-derived trimethylamine may result in interactions with FMO drug substrates immediately after administration; however, the potential for this to occur in vivo may be limited.


Subject(s)
Benzamides/metabolism , Benzyl Compounds/metabolism , Liver/metabolism , Methylamines/metabolism , Oxygenases/metabolism , Animals , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzyl Compounds/chemistry , Benzyl Compounds/pharmacokinetics , Drug Interactions , Liver/chemistry , Male , Methylamines/chemistry , Methylamines/pharmacokinetics , Mice , Mice, Transgenic , Oxygenases/chemistry
5.
Drug Metab Pharmacokinet ; 35(6): 571-573, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32967780

ABSTRACT

Polymorphic human and cynomolgus macaque flavin-containing monooxygenases (FMO) 3 are important oxygenation enzymes for nitrogen-containing drugs. Inter-animal variability of FMO3-dependent drug oxygenations in vivo is suspected in cynomolgus macaques because such variability is evident in humans. Therefore, this follow-up study was performed to investigate the pharmacokinetics of orally administered deuterium-labeled trimethylamine in three cynomolgus macaques genotyped for FMO3. Trimethylamine-d9 was rapidly absorbed and attained plasma concentrations greater than the background levels of non-labeled trimethylamine. Trimethylamine-d9 was then converted to trimethylamine-d9N-oxide. The half-lives, maximum plasma concentrations, and areas under the curve for trimethylamine-d9 and its N-oxygenated metabolite and the total clearance for orally administered trimethylamine-d9 were not different among the heterozygote for Q506K FMO3, the heterozygote for V325I FMO3, and the heterozygote for both S99N and F510S FMO3. Trimethylamine N-oxygenation activities mediated by liver microsomes prepared from the same three animals were not substantially different. However, recombinant proteins of the corresponding cynomolgus FMO3 variants showed apparent reduced trimethylamine N-oxygenation activities compared with the wild-type proteins. This study suggests only limited polymorphic effects on the in vivo catalytic function of cynomolgus FMO3. These findings yield important insights in terms of both quantitative and qualitative variations of polymorphic FMO3 in cynomolgus liver.


Subject(s)
Liver/enzymology , Methylamines/pharmacokinetics , Oxygenases/metabolism , Administration, Oral , Animals , Biotransformation , Genotype , Macaca fascicularis , Male , Methylamines/administration & dosage , Methylamines/blood , Microsomes, Liver/enzymology , Oxidation-Reduction , Oxygenases/genetics , Pharmacogenomic Variants , Phenotype
6.
Article in English | MEDLINE | ID: mdl-32905991

ABSTRACT

N-ethylhexedrone (NEH) and buphedrone (BUPH) are synthetic drugs structurally related to natural cathinone. These synthetic cathinones (SC) are members of the heterogenous family of new psychoactive substances (NPS), which have caused major concern in scientific and forensic communities over the past years, due to their widespread consume. Thus, there is a constant need for monitoring the use of these new substances and gather knowledge on their metabolism and excretion profiles, in order to try to identify markers of NPS consumption. This study aimed at the identification and quantification of NEH, BUPH and selected phase I metabolites using HPLC-MS/MS. NEH, BUPH and some related metabolites were synthesized in-house and quantified in 24 h mice urine, following single dose administration of each drug (64 mg kg-1, i.p.). NEH and BUPH were quantified in mice urine at 58.3 ± 14.4 and 146.2 ± 14.9 µg mL-1, respectively. Similar metabolic pathways were observed for both drugs. Among the metabolites studied, the most excreted ones derived from N-dealkylation of either NEH or BUPH (at around 80 µg mL-1 of urine). Other metabolites resulting from ketone reduction and ketone reduction combined with N-dealkylation or 4-aryl hydroxylation (detected for the first time in non-ring substituted SC) were also identified and quantified. Urine samples were screened using liquid chromatography-high resolution mass spectrometry and various phase II metabolites, including N-acetylated, glucuronides and dicarboxylic acid conjugates were tentatively identified, some of them for the first time. This work is a contribution to the identification of metabolites from SC that can become potential markers to estimate drug consumption.


Subject(s)
Butyrophenones , Chromatography, High Pressure Liquid/methods , Methylamines , Synthetic Drugs , Tandem Mass Spectrometry/methods , Alkaloids , Animals , Butyrophenones/chemistry , Butyrophenones/pharmacokinetics , Butyrophenones/urine , Limit of Detection , Linear Models , Male , Methylamines/chemistry , Methylamines/pharmacokinetics , Methylamines/urine , Mice , Reproducibility of Results , Synthetic Drugs/analysis , Synthetic Drugs/chemistry , Synthetic Drugs/pharmacokinetics
7.
Food Funct ; 11(8): 6745-6776, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32686802

ABSTRACT

There is a growing body of clinical evidence that supports a strong association between elevated circulating trimethylamine N-oxide (TMAO) levels with increased risk of developing adverse cardiovascular outcomes such as atherosclerosis and thrombosis. TMAO is synthesized through a meta-organismal stepwise process that involves (i) the microbial production of TMA in the gut from dietary precursors and (ii) its subsequent oxidation to TMAO by flavin-containing monooxygenases in the liver. Choline, l-carnitine, betaine, and other TMA-containing compounds are the major dietary precursors of TMA. TMAO can also be absorbed directly from the gastrointestinal tract after the intake of TMAO-rich foods such as fish and shellfish. Thus, diet is an important factor as it provides the nutritional precursors to eventually produce TMAO. A number of studies have attempted to associate circulating TMAO levels with the consumption of diets rich in these foods. On the other hand, there is growing interest for the development of novel food ingredients that reduce either the TMAO-induced damage or the endogenous TMAO levels through the interference with microbiota and host metabolic processes involved in TMAO pathway. Such novel functional food ingredients would offer great opportunities to control circulating TMAO levels or its effects, and potentially contribute to decrease cardiovascular risk. In this review we summarize and discuss current data regarding the effects of TMA precursors-enriched foods or diets on circulating TMAO levels, and recent findings regarding the circulating TMAO-lowering effects of specific foods, food constituents and phytochemicals found in herbs, individually or in extracts, and their potential beneficial effect for cardiovascular health.


Subject(s)
Food Ingredients/analysis , Functional Food/analysis , Gastrointestinal Microbiome/physiology , Methylamines/blood , Methylamines/pharmacokinetics , Betaine/metabolism , Biological Availability , Carnitine/metabolism , Choline/metabolism , Diet/methods , Gastrointestinal Absorption , Gastrointestinal Tract/metabolism , Heart Disease Risk Factors , Humans , Liver/metabolism , Oxygenases/metabolism
8.
J Med Chem ; 63(9): 4579-4602, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32282200

ABSTRACT

2-Phenylcyclopropylmethylamine (PCPMA) analogues have been reported as selective serotonin 2C agonists. On the basis of the same scaffold, we designed and synthesized a series of bitopic derivatives as dopamine D3R ligands. A number of these new compounds show a high binding affinity for D3R with excellent selectivity. Compound (1R,2R)-22e and its enantiomer (1S,2S)-22e show a comparable binding affinity for the D3R, but the former is a potent D3R agonist, while the latter acts as an antagonist. Molecular docking studies revealed different binding poses of the PCPMA moiety within the orthosteric binding pocket of the D3R, which might explain the different functional profiles of the enantiomers. Compound (1R,2R)-30q shows a high binding affinity for the D3R (Ki = 2.2 nM) along with good selectivity, as well as good bioavailability and brain penetration properties in mice. These results reveal that the PCPMA scaffold may serve as a privileged scaffold for the design of aminergic GPCR ligands.


Subject(s)
Cyclopropanes/pharmacokinetics , Dopamine Agonists/pharmacokinetics , Dopamine Antagonists/pharmacokinetics , Methylamines/pharmacokinetics , Receptors, Dopamine D3/metabolism , Animals , Binding Sites , Brain/metabolism , Cyclopropanes/chemical synthesis , Cyclopropanes/metabolism , Dopamine Agonists/chemical synthesis , Dopamine Agonists/metabolism , Dopamine Antagonists/chemical synthesis , Dopamine Antagonists/metabolism , Drug Design , Ligands , Methylamines/chemical synthesis , Methylamines/metabolism , Mice, Inbred ICR , Molecular Docking Simulation , Molecular Structure , Receptor, Serotonin, 5-HT2C/metabolism , Stereoisomerism , Structure-Activity Relationship
9.
Toxicol Appl Pharmacol ; 395: 114970, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32234388

ABSTRACT

Synthetic cathinones, such as methylone and pentedrone, are psychoactive derivatives of cathinone, sold in the internet as "plant food" or "bath salts". However, the level at which these compounds and their enantiomers cross the intestinal barrier has not been yet determined. Thus, the present study aimed to analyze the enantioselectivity on the permeability of these drugs through the intestinal barrier by using the Caco-2 cell line, a widely used in vitro model for drug permeability studies. To achieve this goal, an UHPLC-UV method was developed and validated to quantify both synthetic cathinones. The developed UHPLC-UV method revealed high selectivity and a linearity from 1 to 500 µM with correlation coefficients always higher than 0.999. The method has an accuracy that ranged between 89 and 107%, inter-day and intra-day precisions with coefficients of variation below 10%, limits of detection and quantification of 0.31 µM and 0.93 µM for methylone and 0.17 µM and 0.52 µM for pentedrone, respectively. In Caco-2 cells, a differentiated passage of the enantiomers across monolayer was observed for both cathinones. For pentedrone, the difference was observed after the first hour, being R-(-)-pentedrone the most permeable compound. Regarding methylone, the difference was noted after one hour and 30 min, with S-(-)-methylone being the most absorbed enantiomer. In conclusion, a fully validated method was successfully applied for studying the permeability of methylone and pentedrone enantiomers in an in vitro model of human intestine, which allowed to discover, for the first time, the enantioselectivity in drug permeability of this class of drugs.


Subject(s)
Chromatography, High Pressure Liquid/methods , Intestinal Absorption/physiology , Methamphetamine/analogs & derivatives , Methylamines/chemistry , Methylamines/pharmacokinetics , Pentanones/chemistry , Pentanones/pharmacokinetics , Alkaloids/chemistry , Caco-2 Cells , Humans , Methamphetamine/chemistry , Methamphetamine/pharmacokinetics , Permeability , Psychotropic Drugs , Sensitivity and Specificity , Stereoisomerism , Structure-Activity Relationship
10.
J Pharm Biomed Anal ; 170: 176-186, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30927663

ABSTRACT

In this study, a simple, fast and sensitive LC/MS/MS method was developed and validated for the determination of GW9508 in rat plasma. The sample was precipitated with acetonitrile and subsequently separated on ZORBAX Eclipse XDB C18 column (50 mm × 2.1 mm, 5 µm). Mobile phase was composed of 0.1% formic acid in water and acetonitrile with gradient elution, at a flow rate of 0.4 mL/min. The analyte and internal standard were quantitatively monitored with precursor-to-product transitions of m/z 348.2→183.1 and m/z 397.2→260.2, respectively. The linearity of the assay was evident in the range of 1-1000 ng/mL with correlation coefficient more than 0.998. The validation parameters were all within the acceptable limits. The validated method has been successfully applied to the pharmacokinetics study of GW9508 in rat plasma, and our results demonstrated that GW9508 showed low clearance, moderate half-life and ideal bioavailability (54.88%). Furthermore, metabolites stemmed from rat plasma, rat hepatocytes and human hepatocytes were analyzed by an LC-Q-Exactive-Orbitrap-MS assay, resulting in the identification of seven metabolites based on the accurate mass and fragment ions. Acylglucuronide conjugate (M6) was found as the most abundant metabolite in all tested matrices. The metabolic pathways were proposed as hydroxylation and glucuronidation. This study provided an overview of disposition of GW9508, which is highly instructive for better understanding the effectiveness and toxicity of this drug.


Subject(s)
Methylamines/metabolism , Methylamines/pharmacokinetics , Propionates/metabolism , Propionates/pharmacokinetics , Acetonitriles/pharmacokinetics , Animals , Biological Availability , Chromatography, Liquid/methods , Hepatocytes/metabolism , Humans , Male , Plasma/metabolism , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods
11.
Article in English | MEDLINE | ID: mdl-30670430

ABSTRACT

DS86760016 is a new leucyl-tRNA-synthetase inhibitor at the preclinical development stage. DS86760016 showed potent activity against extended-spectrum multidrug-resistant Pseudomonas aeruginosa isolated from clinical samples and in vitro biofilms. In a murine catheter-associated urinary tract infection model, DS86760016 treatment resulted in significant eradication of P. aeruginosa from the kidney, bladder, and catheter without developing drug resistance. Our data suggest that DS86760016 has the potential to act as a new drug for the treatment of Pseudomonas infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Boron Compounds/pharmacology , Catheter-Related Infections/drug therapy , Dioxoles/pharmacology , Leucine-tRNA Ligase/antagonists & inhibitors , Methylamines/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Urinary Tract Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Biofilms/growth & development , Boron Compounds/pharmacokinetics , Catheter-Related Infections/microbiology , Dioxoles/pharmacokinetics , Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Female , Humans , Methylamines/pharmacokinetics , Mice , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Urinary Tract Infections/microbiology
12.
Gut ; 68(8): 1439-1449, 2019 08.
Article in English | MEDLINE | ID: mdl-30377191

ABSTRACT

OBJECTIVE: The gut microbiota-derived metabolite, trimethylamine N-oxide (TMAO) plays an important role in cardiovascular disease (CVD). The fasting plasma TMAO was shown as a prognostic indicator of CVD incident in patients and raised the interest of intervention targeting gut microbiota. Here we develop a clinically applicable method called oral carnitine challenge test (OCCT) for TMAO-related therapeutic drug efforts assessment and personalising dietary guidance. DESIGN: A pharmacokinetic study was performed to verify the design of OCCT protocol. The OCCT was conducted in 23 vegetarians and 34 omnivores to validate gut microbiota TMAO production capacity. The OCCT survey was integrated with gut microbiome, host genotypes, dietary records and serum biochemistry. A humanised gnotobiotic mice study was performed for translational validation. RESULTS: The OCCT showed better efficacy than fasting plasma TMAO to identify TMAO producer phenotype. The omnivores exhibited a 10-fold higher OR to be high TMAO producer than vegetarians. The TMAO-associated taxa found by OCCT in this study were consistent with previous animal studies. The TMAO producer phenotypes were also reproduced in humanised gnotobiotic mice model. Besides, we found the faecal CntA gene was not associated with TMAO production; therefore, other key relevant microbial genes might be involved. Finally, we demonstrated the urine TMAO exhibited a strong positive correlation with plasma TMAO (r=0.92, p<0.0001) and improved the feasibility of OCCT. CONCLUSION: The OCCT can be used to identify TMAO-producer phenotype of gut microbiota and may serve as a personal guidance in CVD prevention and treatment. TRIAL REGISTRATION NUMBER: NCT02838732; Results.


Subject(s)
Carnitine/pharmacology , Dysbiosis , Feeding Behavior/physiology , Gastrointestinal Microbiome/physiology , Methylamines , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Carnitine/metabolism , Diet/methods , Dysbiosis/diagnosis , Dysbiosis/metabolism , Humans , Methylamines/metabolism , Methylamines/pharmacokinetics , Mice , Oxidants/metabolism , Oxidants/pharmacokinetics , Prognosis , Renal Elimination/physiology
13.
Clin Pharmacol Drug Dev ; 8(1): 70-77, 2019 01.
Article in English | MEDLINE | ID: mdl-29870578

ABSTRACT

GSK2330672 is an inhibitor of the ileal bile acid transporter, designed to have minimal systemic exposure, and is under development as a potential therapeutic for pruritus associated with primary biliary cholangitis and other cholestatic liver diseases. A phase 1, double-blind, placebo-controlled, 4-period crossover study was conducted to evaluate the safety, tolerability, and pharmacokinetic/pharmacodynamic characteristics of GSK2330672 in healthy Japanese participants. Sixteen healthy male participants received single oral doses of GSK2330672 (10-180 mg) or placebo in each period. No serious adverse events and no adverse events leading to study discontinuation or withdrawal were reported. Drug-related adverse events reported included gastrointestinal symptoms (mostly diarrhea) and positive fecal occult blood tests, and were all mild and resolved without any interventions. GSK2330672 was undetectable in the majority of participants' plasma. Pharmacodynamic observations included a tendency for total serum bile acids to reduce and for serum 7α-hydroxy-4-cholesten-3-one, a key intermediate of bile acid synthesis, to increase with increasing doses of GSK2330672. In the context of recently published indications of potential efficacy for cholestatic pruritus in non-Japanese populations, these data support further evaluations of GSK2330672 in Japanese patients.


Subject(s)
Methylamines/pharmacology , Methylamines/pharmacokinetics , Thiazepines/pharmacology , Thiazepines/pharmacokinetics , Adult , Asian People , Carrier Proteins/antagonists & inhibitors , Cholestenones/blood , Cross-Over Studies , Double-Blind Method , Healthy Volunteers , Humans , Male , Membrane Glycoproteins/antagonists & inhibitors , Methylamines/adverse effects , Methylamines/blood , Middle Aged , Thiazepines/adverse effects , Thiazepines/blood , Young Adult
14.
J Toxicol Sci ; 43(6): 387-393, 2018.
Article in English | MEDLINE | ID: mdl-29877215

ABSTRACT

Medicinal carnitine-derived and dietary-derived malodorous trimethylamine and its non-malodorous metabolite trimethylamine N-oxide were historically regarded as nontoxic. Clinical and toxicological interest has recently arisen because of their potential association with atherosclerosis. We previously reported a human physiologically based pharmacokinetic (PBPK) model for trimethylamine and its primary metabolite, trimethylamine N-oxide, based on reported rat trimethylamine pharmacokinetics. However, rats are poor metabolizers with respect to trimethylamine N-oxygenation, and this species difference was investigated in vitro using substrate depletion rates in rat and human liver microsomes. The current study investigated the pharmacokinetics of deuterium-labeled trimethylamine orally administered to immunodeficient humanized-liver mice transplanted with commercially available human hepatocytes. Trimethylamine N-oxide was extensively formed in vivo in humanized-liver mice, but not in control mice. The experimental pharmacokinetic data of deuterium-labeled trimethylamine and its N-oxide in humanized-liver mice were scaled up for application to a human PBPK model. The human plasma concentration curves generated by the resulting simple PBPK model were consistent with concentrations in humans reported in the literature. The model can also simulate human plasma levels of trimethylamine and trimethylamine N-oxide during treatment with the prescription medicine L-carnitine and in trimethylamine loading tests. The predicted plasma levels were in the ranges that occur under the consumption of daily dietary foodstuff; such levels are associated with few toxicological impacts. The present PBPK model for trimethylamine and trimethylamine N-oxide could estimate daily doses by both forward and reverse dosimetry and could facilitate risk assessment in humans.


Subject(s)
Methylamines/administration & dosage , Methylamines/pharmacokinetics , Microsomes, Liver/metabolism , Administration, Oral , Animals , Atherosclerosis/chemically induced , Deuterium , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Methylamines/blood , Methylamines/toxicity , Mice , Models, Animal , Models, Biological , Rats , Risk Assessment
15.
Chem Biol Drug Des ; 92(2): 1547-1554, 2018 08.
Article in English | MEDLINE | ID: mdl-29777569

ABSTRACT

The free fatty acid receptor 1 (FFA1) is a potential target due to its function in enhancing of glucose-stimulated insulin secretion. The FFA1 agonist GW9508 has great potential for the treatment of type 2 diabetes mellitus, but it has been suffering from high plasma clearance probably because the phenylpropanoic acid is vulnerable to ß-oxidation. To identify orally available analog without influence on the unique pharmacological mechanism of GW9508, we tried to interdict the metabolically labile group by incorporating two deuterium atoms at the α-position of phenylpropionic acid affording compound 4 (HWL-066). As expected, HWL-066 revealed a lower clearance (CL = 0.23 L-1  hr-1  kg-1 ), higher maximum concentration (Cmax  = 5907.47 µg/L), and longer half-life (T1/2  = 3.50 hr), resulting in a 2.8-fold higher exposure than GW9508. Moreover, the glucose-lowering effect of HWL-066 was far better than that of GW9508 and comparable with TAK-875. Different from glibenclamide, no side-effect of hypoglycemia was observed in mice after oral administrating HWL-066 (80 mg/kg).


Subject(s)
Hypoglycemic Agents/chemistry , Propionates/chemistry , Receptors, G-Protein-Coupled/agonists , Animals , Binding Sites , Calcium/metabolism , Deuterium/chemistry , Deuterium/metabolism , Glucose Tolerance Test , Half-Life , Humans , Hydrogen Bonding , Hypoglycemia/drug therapy , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Male , Methylamines/pharmacokinetics , Methylamines/pharmacology , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Propionates/metabolism , Propionates/pharmacokinetics , Propionates/pharmacology , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
16.
J Toxicol Sci ; 42(4): 485-490, 2017.
Article in English | MEDLINE | ID: mdl-28717107

ABSTRACT

Trimethylamine, a dietary- and medicinal carnitine-derived amine, is extensively metabolized by liver to non-malodorous trimethylamine N-oxide. Although trimethylamine and trimethylamine N-oxide under daily dietary consumption or carnitine treatment are generally regarded as nontoxic, they have been, and remain, of toxicological and clinical interest because of their potential association with atherosclerosis. The aim of the current study was to model the pharmacokinetics of trimethylamine after oral administration of trimethylamine in humans and compare the results with reported measured values. Adjusted biomonitoring equivalents from rat studies based on reported plasma concentrations were scaled to human equivalents using known species allometric scaling factors. In vitro metabolic clearance data were obtained using rat and human liver microsomal preparations. Renal clearances in humans for trimethylamine and trimethylamine N-oxide were calculated with a clearance concept approach using reported 24-hr urinary excretion rates and assumed areas under plasma concentration curves. The resulting modeled plasma and urinary concentration curves by simple physiologically based pharmacokinetic models (or semi-physiological pharmacokinetic models) were consistent with reported concentrations. This study provides important information to help simulate human plasma levels of trimethylamine and trimethylamine N-oxide in trimethylamine loading tests and during treatment with prescribed medicinal l-carnitine, showing the similar range as that resulting from daily dietary foodstuff consumption along with little toxicological impacts. The present models could estimate relationship between plasma and urine concentrations of trimethylamine or trimethylamine N-oxide and the daily oral doses by both forward and reverse dosimetry from viewpoint of human risk assessment.


Subject(s)
Methylamines/blood , Methylamines/urine , Models, Biological , Administration, Oral , Animals , Carnitine , Humans , In Vitro Techniques , Kidney/metabolism , Male , Methylamines/administration & dosage , Methylamines/pharmacokinetics , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Risk Assessment
17.
J Nutr Biochem ; 45: 77-82, 2017 07.
Article in English | MEDLINE | ID: mdl-28433924

ABSTRACT

Trimethylamine-N-oxide (TMAO) is associated with chronic disease risk. However, little is known about the metabolic fate of dietary TMAO. This study sought to quantitatively elucidate the metabolic fate of orally consumed TMAO in humans. As part of a crossover feeding study, healthy young men (n=40) consumed 50-mg deuterium-labeled methyl d9-TMAO (d9-TMAO), and enrichments of TMAO and its derivatives were measured in blood for 6 h, urine and stool, as well as skeletal muscle in a subset of men (n=6). Plasma d9-TMAO was detected as early as 15 min, increased until 1 h and remained elevated through the 6-h period. TMAO exhibited an estimated turnover time of 5.3 h, and ~96% of the dose was eliminated in urine by 24 h, mainly as d9-TMAO. No d9-TMAO was detected in feces. Notably, d9-TMAO and d9-trimethylamine were detected in skeletal muscle (n=6) at 6 h, and the enrichment ratio of d9-TMAO to d9-trimethylamine was influenced by a genetic variant in flavin-containing monooxygenase isoform 3 (FMO3 G472A). These results suggest that the absorption of orally consumed TMAO is near complete and does not require processing by gut microbes. TMAO exhibits fast turnover in the circulation with the majority being eliminated in urine within 24 h. A small portion of the dose, however, is taken up by extrahepatic tissue in a manner that appears to be under the influence of FMO3 G472A polymorphism. This trial was registered at clinicaltrials.gov as NCT02558673.


Subject(s)
Methylamines/pharmacokinetics , Oxygenases/genetics , Administration, Oral , Adult , Deuterium , Humans , Male , Methylamines/administration & dosage , Methylamines/blood , Methylamines/urine , Middle Aged , Muscle, Skeletal/metabolism , Oxygenases/metabolism , Polymorphism, Genetic
18.
BMC Gastroenterol ; 16(1): 71, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27431238

ABSTRACT

BACKGROUND: Pruritus (itch) is a symptom commonly experienced by patients with cholestatic liver diseases such as primary biliary cholangitis (PBC, previously referred to as primary biliary cirrhosis). Bile acids (BAs) have been proposed as potential pruritogens in PBC. The ileal bile acid transporter (IBAT) protein expressed in the distal ileum plays a key role in the enterohepatic circulation of BAs. Pharmacological inhibition of IBAT with GSK2330672 may reduce BA levels in the systemic circulation and improve pruritus. METHODS: This clinical study (BAT117213 study) is sponsored by GlaxoSmithKline (GSK) with associated exploratory studies supported by the National Institute for Health Research (NIHR). It is a phase 2a, multi-centre, randomised, double bind, placebo controlled, cross-over trial for PBC patients with pruritus. The primary objective is to investigate the safety and tolerability of repeat doses of GSK2330672, and explore whether GSK2330672 administration for 14 days improves pruritus compared with placebo. The key outcomes include improvement in pruritus scores evaluated on a numerical rating scale and other PBC symptoms in an electronic diary completed twice daily by the patients. The secondary outcomes include the evaluation of the effect of GSK2330672 on total serum bile acid (BA) concentrations, serum markers of BA synthesis and steady-state pharmacokinetics of ursodeoxycholic acid (UDCA). DISCUSSION: BAT117213 study is the first randomised controlled crossover trial of ileal bile acid transporter inhibitor, a novel class of drug to treat pruritus in PBC. The main strengths of the trial are utility of a novel, study specific, electronic symptom diary as patient reported outcome to measure the treatment response objectively and the crossover design that allows estimating the treatment effect in a smaller number of patients. The outcome of this trial will inform the trial design of future development phase of the IBAT inhibitor drug. The trial will also provide opportunity to conduct metabonomic and gut microbiome studies as explorative and mechanistic research in patients with cholestatic pruritus. TRIAL REGISTRATION: EudraCT number: 2012-005531-84, ClinicalTrials.gov Identifier: NCT01899703 , registered on 3(rd) July 2013.


Subject(s)
Liver Cirrhosis, Biliary/complications , Methylamines/therapeutic use , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Pruritus/drug therapy , Symporters/antagonists & inhibitors , Thiazepines/therapeutic use , Adolescent , Adult , Aged , Bile Acids and Salts/blood , Biomarkers/blood , Cholagogues and Choleretics/pharmacokinetics , Cholagogues and Choleretics/therapeutic use , Cross-Over Studies , Drug Therapy, Combination , Female , Humans , Male , Methylamines/administration & dosage , Methylamines/adverse effects , Methylamines/pharmacokinetics , Middle Aged , Organic Anion Transporters, Sodium-Dependent/therapeutic use , Pruritus/etiology , Symporters/therapeutic use , Thiazepines/administration & dosage , Thiazepines/adverse effects , Thiazepines/pharmacokinetics , Ursodeoxycholic Acid/pharmacokinetics , Ursodeoxycholic Acid/therapeutic use , Young Adult
19.
J Pharmacokinet Pharmacodyn ; 42(6): 591-609, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26194069

ABSTRACT

Any statistical model should be identifiable in order for estimates and tests using it to be meaningful. We consider statistical analysis of physiologically-based pharmacokinetic (PBPK) models in which parameters cannot be estimated precisely from available data, and discuss different types of identifiability that occur in PBPK models and give reasons why they occur. We particularly focus on how the mathematical structure of a PBPK model and lack of appropriate data can lead to statistical models in which it is impossible to estimate at least some parameters precisely. Methods are reviewed which can determine whether a purely linear PBPK model is globally identifiable. We propose a theorem which determines when identifiability at a set of finite and specific values of the mathematical PBPK model (global discete identifiability) implies identifiability of the statistical model. However, we are unable to establish conditions that imply global discrete identifiability, and conclude that the only safe approach to analysis of PBPK models involves Bayesian analysis with truncated priors. Finally, computational issues regarding posterior simulations of PBPK models are discussed. The methodology is very general and can be applied to numerous PBPK models which can be expressed as linear time-invariant systems. A real data set of a PBPK model for exposure to dimethyl arsinic acid (DMA(V)) is presented to illustrate the proposed methodology.


Subject(s)
Cacodylic Acid/pharmacokinetics , Environmental Exposure , Environmental Pollutants/pharmacokinetics , Models, Biological , Models, Statistical , Animals , Bayes Theorem , Biotransformation , Cacodylic Acid/adverse effects , Cacodylic Acid/urine , Computer Simulation , Environmental Exposure/adverse effects , Environmental Pollutants/adverse effects , Environmental Pollutants/urine , Humans , Linear Models , Methylamines/pharmacokinetics , Mice , Risk Assessment
20.
Article in English | MEDLINE | ID: mdl-25545914

ABSTRACT

In vitro gut sac experiments were performed on freshwater and 60% seawater acclimated trout (Oncorhynchus mykiss) under treatments designed to discern possible mechanisms of intestinal ammonia transport. Seawater acclimation increased ammonia flux rate into the serosal saline (Jsamm) in the anterior intestine, however it did not alter Jsamm in the mid- or posterior intestine suggesting similar mechanisms of ammonia handling in freshwater and seawater fish. Both fluid transport rate (FTR) and Jsamm were inhibited in response to basolateral ouabain treatment, suggesting a linkage of ammonia uptake to active transport, possibly coupled to fluid transport processes via solvent drag. Furthermore, decreases in FTR and Jsamm caused by low Na(+) treatment indicated a Na(+) linked transport mechanism. Mucosal bumetanide (10(-4) M) had no impact on FTR, yet decreased Jsamm in the anterior and mid-intestine, suggesting NH4(+) substitution for K(+) on an apical NKCC, and at least a partial uncoupling of ammonia transport from fluid transport. Additional treatments (amiloride, 5-(N-ethyl-N-isopropyl)amiloride (EIPA), phenamil, bafilomycin, 4',6-diamidino-2-phenylindole (DAPI), high sodium) intended to disrupt alternative routes of Na(+) uptake yielded no change in FTR or Jsamm, suggesting the absence of direct competition between Na(+) and ammonia for transport. Finally, [(14)C]methylamine permeability (PMA) measurements indicated the likely presence of an intestinal Rh-mediated ammonia transport system, as increasing NH4Cl (0, 1, 5 mmol l(-1)) concentrations reduced PMA, suggesting competition for transport through Rh proteins. Overall, the data presented in this paper provide some of the first insights into mechanisms of teleost intestinal ammonia transport.


Subject(s)
Acclimatization/physiology , Ammonia/metabolism , Intestinal Mucosa/metabolism , Oncorhynchus mykiss/physiology , Sodium/metabolism , Amiloride/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/physiology , Bumetanide/pharmacology , Fresh Water , Intestines/drug effects , Methylamines/metabolism , Methylamines/pharmacokinetics , Oncorhynchus mykiss/metabolism , Organ Culture Techniques/methods , Permeability , Seawater , Sodium Potassium Chloride Symporter Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...