29 research outputs found

    Metabolic Activation of Fluoropyrrolidine Dipeptidyl Peptidase-IV Inhibitors by Rat Liver Microsomes

    Get PDF
    Abstract The current study evaluated the potential for two dipeptidyl peptidase-IV (DPP-IV) inhibitor analogues (MRL-A and MRL-B), containing a fluoropyrrolidine moiety in the structure, to undergo metabolic activation. The irreversible binding of these tritiumlabeled compounds to rat liver microsomal protein was time-and NADPH-dependent, and was attenuated by the addition of reduced glutathione (GSH) or N-acetylcysteine (NAC) to the incubation, indicating that chemically reactive intermediates were formed and trapped by these nucleophiles. Mass spectrometric analyses and further trapping experiments with semicarbazide indicated that the fluoropyrrolidine ring had undergone sequential oxidation and defluorination events resulting in the formation of GSH or NAC conjugates of the pyrrolidine moiety. The bioactivation of MRL-A was catalyzed primarily by rat recombinant cytochrome (CYP) 3A1 and 3A2. Pretreatment of rats with prototypic CYP3A1 and 3A2 inducers (pregnenolone-16alpha-carbonitrile (PCN) and dexamethasone) enhanced the extent of bioactivation, which in turn, led to a higher degree of in vitro irreversible binding to microsomal proteins (5-and 9-fold increase, respectively). Herein, we describe studies which demonstrate that the fluoropyrrolidine ring is prone to metabolic activation, and that GSH or NAC can trap the reactive intermediates to form adducts that provide insight into the mechanisms of bioactivation

    Determination of acetyl coenzyme A in human whole blood by ultra-performance liquid chromatography-mass spectrometry

    No full text
    Acetyl coenzyme A is involved in several key metabolic pathways. Its concentration can vary considerably in response to physiological or pathological conditions making it a potentially valuable biomarker. However, little information about the measurement and concentration of acetyl CoA in human whole blood is found in the literature. The aim of this study was the development of an accurate method for the determination of acetyl CoA in human whole blood by LC-MS/MS. The method, involving extraction from whole blood by a rapid protein precipitation procedure was thoroughly validated: limit of quantitation was 3.91 ng mL-1. Accuracy and precision were calculated at five concentrations and were within ±15%. The average endogenous level of acetyl CoA in human whole blood was determined in 17 healthy individuals to be 220.9 ng mL-1 (ranging from 124.0 to 308.0 ng mL-1). This represents, to our knowledge, the first report of acetyl CoA levels in human whole blood, and the first practical and reliable method for its determination

    Pharmacokinetics of memantine in rats and mice

    No full text
    To evaluate the potential of memantine as a therapeutic agent for Huntington’s disease (HD) we have undertaken a series of in vitro, ex vivo and whole animal studies to characterize its pharmacokinetics (PK) and pharmacodynamics (PD) in rats and mice. Results from these studies will enable determination of memantine exposures needed to engage the related functional PD marker and help predict the dose regimen for clinical trials to test its proposed mechanism of action; the selective blockade of extrasynaptic, but not synaptic, NMDA receptors. The studies reported here describe the PK of memantine in rats and mice at low (1 mg/kg) and high (10 mg/kg) doses. Our studies indicate that the clearance mechanisms of memantine in rats and mice are different from those in human, and that clearance needs to be taken into account when extrapolating to the human. In rats only, there is a significant metabolic contribution to memantine clearance at lower dose levels. While memantine is primarily cleared renally in all three species, the proportion of total systemic clearance above the glomerular filtration rate (GFR) is much higher in rats and mice (~13, 4.5, and 1.4 times higher than GFR in rats, mice, and humans, respectively), suggesting that the contribution of active transport to memantine elimination in rats and mice is more significant than in the human. In rats and mice, memantine had a short half-life (<4 h) and steep Cmax/Cmin ratios (>100). In the human, the half-life of memantine was reported to be very long (60-80 h) with a Cmax/Cmin ratio at steady state concentrations of ~1.5. A small change in the clearance of memantine - for example due to renal impairment or competition for the elimination pathway with a co-administered drug - will likely affect exposure and, therefore, the selectivity of memantine on NMDA receptors . The PK differences observed between these species demonstrate that the PK in mice and rats cannot be directly extrapolated to the human. Further, the relationship between the plasma concentration (and therefore dose) needed to elicit a mechanism-related in vivo functional effect (PD readout) while maintaining the selectivity of the extrasynaptic blockade of the NMDA receptors needs to be established before clinical trials can be appropriately planned

    Metabolism of [ 14

    No full text

    Fosmetpantotenate (RE-024), a phosphopantothenate replacement therapy for pantothenate kinase-associated neurodegeneration: Mechanism of action and efficacy in nonclinical models

    No full text
    <div><p>In cells, phosphorylation of pantothenic acid to generate phosphopantothenic acid by the pantothenate kinase enzymes is the first step in coenzyme A synthesis. Pantothenate kinase 2, the isoform localized in neuronal cell mitochondria, is dysfunctional in patients with pantothenate kinase-associated neurodegeneration. Fosmetpantotenate is a phosphopantothenic acid prodrug in clinical development for treatment of pantothenate kinase-associated neurodegeneration, which aims to replenish phosphopantothenic acid in patients. Fosmetpantotenate restored coenzyme A in short-hairpin RNA pantothenate kinase 2 gene-silenced neuroblastoma cells and was permeable in a blood-brain barrier model. The rate of fosmetpantotenate metabolism in blood is species-dependent. Following up to 700 mg/kg orally, blood exposure to fosmetpantotenate was negligible in rat and mouse, but measurable in monkey. Consistent with the difference in whole blood half-life, fosmetpantotenate dosed orally was found in the brains of the monkey (striatal dialysate) but was absent in mice. Following administration of isotopically labeled-fosmetpantotenate to mice, ~40% of liver coenzyme A (after 500 mg/kg orally) and ~50% of brain coenzyme A (after 125 μg intrastriatally) originated from isotopically labeled-fosmetpantotenate. Additionally, 10-day dosing of isotopically labeled-fosmetpantotenate, 12.5 μg, intracerebroventricularly in mice led to ~30% of brain coenzyme A containing the stable isotopic labels. This work supports the hypothesis that fosmetpantotenate acts to replace reduced phosphopantothenic acid in pantothenate kinase 2-deficient tissues.</p></div

    Mean half—Life of fosmetpantotenate and diastereomers after incubation with blood from various species at 37°C for 60 min<sup>a</sup>.

    No full text
    <p>Mean half—Life of fosmetpantotenate and diastereomers after incubation with blood from various species at 37°C for 60 min<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0192028#t002fn001" target="_blank"><sup>a</sup></a>.</p

    Effects of 1 μM fosmetpantotenate TID for 5 consecutive days in shRNA PanK2 knockdown human neuroblastoma cells.

    No full text
    <p>(A) Intracellular CoA concentrations, n = 3. (B) Western blot densitometry values. β-actin was used for normalization. Two experiments in duplicate. Two sided t-test; *p ≤0.05, **p ≤0.01, ***p ≤0.001. Gel images can be found in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0192028#pone.0192028.s002" target="_blank">S2 Fig</a>.</p

    Fosmetpantotenate, PPA, and total PA in monkey blood.

    No full text
    <p>Concentrations of fosmetpantotenate, PPA, and PA after a single oral administration of fosmetpantotenate in cynomolgus monkeys at 300 mg/kg (N = 2).</p
    corecore