Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria Beconi is active.

Publication


Featured researches published by Maria Beconi.


Bioorganic & Medicinal Chemistry Letters | 2008

Fluoroolefins as amide bond mimics in dipeptidyl peptidase IV inhibitors

Scott D. Edmondson; Lan Wei; Jinyou Xu; Jackie Shang; Shiyao Xu; Jianmei Pang; Ashok Chaudhary; Dennis C. Dean; Huaibing He; Barbara Leiting; Kathryn A. Lyons; Reshma A. Patel; Sangita B. Patel; Giovanna Scapin; Joseph K. Wu; Maria Beconi; Nancy A. Thornberry; Ann E. Weber

The synthesis, selectivity, rat pharmacokinetic profile, and drug metabolism profiles of a series of potent fluoroolefin-derived DPP-4 inhibitors (4) are reported. A radiolabeled fluoroolefin 33 was shown to possess a high propensity to form reactive metabolites, thus revealing a potential liability for this class of DPP-4 inhibitors.


Drug Metabolism and Disposition | 2007

Disposition of the dipeptidyl peptidase 4 inhibitor sitagliptin in rats and dogs.

Maria Beconi; James R. Reed; Yohannes Teffera; Yuan Qing Xia; Christopher J. Kochansky; David Q. Liu; Shiyao Xu; Charles S. Elmore; Suzanne L. Ciccotto; Donald F. Hora; Ralph A. Stearns; Stella H. Vincent

The pharmacokinetics, metabolism, and excretion of sitagliptin [MK-0431; (2R)-4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5-trifluorophenyl)butan-2-amine], a potent dipeptidyl peptidase 4 inhibitor, were evaluated in male Sprague-Dawley rats and beagle dogs. The plasma clearance and volume of distribution of sitagliptin were higher in rats (40–48 ml/min/kg, 7–9 l/kg) than in dogs (∼9 ml/min/kg, ∼3 l/kg), and its half-life was shorter in rats, ∼2 h compared with ∼4 h in dogs. Sitagliptin was absorbed rapidly after oral administration of a solution of the phosphate salt. The absolute oral bioavailability was high, and the pharmacokinetics were fairly dose-proportional. After administration of [14C]sitagliptin, parent drug was the major radioactive component in rat and dog plasma, urine, bile, and feces. Sitagliptin was eliminated primarily by renal excretion of parent drug; biliary excretion was an important pathway in rats, whereas metabolism was minimal in both species in vitro and in vivo. Approximately 10 to 16% of the radiolabeled dose was recovered in the rat and dog excreta as phase I and II metabolites, which were formed by N-sulfation, N-carbamoyl glucuronidation, hydroxylation of the triazolopiperazine ring, and oxidative desaturation of the piperazine ring followed by cyclization via the primary amine. The renal clearance of unbound drug in rats, 32 to 39 ml/min/kg, far exceeded the glomerular filtration rate, indicative of active renal elimination of parent drug.


Journal of Medicinal Chemistry | 2012

Discovery and Structure–Activity Relationship of Potent and Selective Covalent Inhibitors of Transglutaminase 2 for Huntington’s Disease

Michael Prime; Ole Andreas Andersen; John J. Barker; Mark Brooks; Robert K. Y. Cheng; Ian Toogood-Johnson; Stephen Martin Courtney; Frederick Arthur Brookfield; Christopher John Yarnold; Richard W. Marston; Peter Johnson; Siw Johnsen; Jordan J. Palfrey; Darshan Vaidya; Sayeh Erfan; Osamu Ichihara; Brunella Felicetti; Shilpa Palan; Anna Pedret-Dunn; Sabine Schaertl; Ina Sternberger; Andreas Ebneth; Andreas Scheel; Dirk Winkler; Leticia Toledo-Sherman; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez; John Wityak

Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntingtons disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.


Journal of Medicinal Chemistry | 2015

Development of a Series of Aryl Pyrimidine Kynurenine Monooxygenase Inhibitors as Potential Therapeutic Agents for the Treatment of Huntington's Disease

Leticia Toledo-Sherman; Michael Prime; Ladislav Mrzljak; Maria Beconi; Alan Beresford; Frederick Arthur Brookfield; Christopher John Brown; Isabell Cardaun; Stephen Martin Courtney; Ulrike Dijkman; Estelle Hamelin-Flegg; Peter Johnson; Valerie Kempf; Kathy Lyons; Kimberly Matthews; William Leonard Mitchell; Catherine O’Connell; Paula Pena; Kendall Powell; Arash Rassoulpour; Laura Reed; Wolfgang Reindl; Suganathan Selvaratnam; Weslyn Ward Friley; Derek Weddell; Naomi Went; Patricia Wheelan; Christin Winkler; Dirk Winkler; John Wityak

We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.


ACS Medicinal Chemistry Letters | 2012

SAR Development of Lysine-Based Irreversible Inhibitors of Transglutaminase 2 for Huntington's Disease

John Wityak; Michael Prime; Frederick Arthur Brookfield; Stephen Martin Courtney; Sayeh Erfan; Siw Johnsen; Peter Johnson; Marie Li; Richard W. Marston; Laura Reed; Darshan Vaidya; Sabine Schaertl; Anna Pedret-Dunn; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez

We report a series of irreversible transglutaminase 2 inhibitors starting from a known lysine dipeptide bearing an acrylamide warhead. We established new SARs resulting in compounds demonstrating improved potency and better physical and calculated properties. Transglutaminase selectivity profiling and in vitro ADME properties of selected compounds are also reported.


Journal of Pharmaceutical and Biomedical Analysis | 2015

A single-run liquid chromatography mass spectrometry method to quantify neuroactive kynurenine pathway metabolites in rat plasma.

Laura Orsatti; Roberto Speziale; Maria Vittoria Orsale; Fulvia Caretti; Maria Veneziano; Matteo Zini; Edith Monteagudo; Kathryn A. Lyons; Maria Beconi; Kelvin Chan; Todd Herbst; Leticia Toledo-Sherman; Ignacio Munoz-Sanjuan; Fabio Bonelli; Celia Dominguez

Neuroactive metabolites in the kynurenine pathway of tryptophan catabolism are associated with neurodegenerative disorders. Tryptophan is transported across the blood-brain barrier and converted via the kynurenine pathway to N-formyl-L-kynurenine, which is further degraded to L-kynurenine. This metabolite can then generate a group of metabolites called kynurenines, most of which have neuroactive properties. The association of tryptophan catabolic pathway alterations with various central nervous system (CNS) pathologies has raised interest in analytical methods to accurately quantify kynurenines in body fluids. We here describe a rapid and sensitive reverse-phase HPLC-MS/MS method to quantify L-kynurenine (KYN), kynurenic acid (KYNA), 3-hydroxy-L-kynurenine (3HK) and anthranilic acid (AA) in rat plasma. Our goal was to quantify these metabolites in a single run; given their different physico-chemical properties, major efforts were devoted to develop a chromatography suitable for all metabolites that involves plasma protein precipitation with acetonitrile followed by chromatographic separation by C18 RP chromatography, detected by electrospray mass spectrometry. Quantitation range was 0.098-100 ng/ml for 3HK, 9.8-20,000 ng/ml for KYN, 0.49-1000 ng/ml for KYNA and AA. The method was linear (r>0.9963) and validation parameters were within acceptance range (calibration standards and QC accuracy within ±30%).


Journal of Medicinal Chemistry | 2015

Lead Optimization toward Proof-of-Concept Tools for Huntington’s Disease within a 4-(1H-Pyrazol-4-yl)pyrimidine Class of Pan-JNK Inhibitors

John Wityak; Kevin McGee; Michael Conlon; Ren Hua Song; Bryan Cordell Duffy; Brent Clayton; Michael P. Lynch; Gwen Wang; Emily Freeman; James C. Haber; Douglas B. Kitchen; David D. Manning; Jiffry Ismail; Yuri L. Khmelnitsky; Peter C. Michels; Jeff Webster; Macarena Irigoyen; Michele Luche; Monica Hultman; Mei Bai; IokTeng D. Kuok; Ryan Newell; Marieke Lamers; Philip M. Leonard; Dawn Yates; Kim L. Matthews; Lynette Ongeri; Steve Clifton; Tania Mead; Susan Deupree

Through medicinal chemistry lead optimization studies focused on calculated properties and guided by X-ray crystallography and computational modeling, potent pan-JNK inhibitors were identified that showed submicromolar activity in a cellular assay. Using in vitro ADME profiling data, 9t was identified as possessing favorable permeability and a low potential for efflux, but it was rapidly cleared in liver microsomal incubations. In a mouse pharmacokinetics study, compound 9t was brain-penetrant after oral dosing, but exposure was limited by high plasma clearance. Brain exposure at a level expected to support modulation of a pharmacodynamic marker in mouse was achieved when the compound was coadministered with the pan-cytochrome P450 inhibitor 1-aminobenzotriazole.


Xenobiotica | 2005

Metabolic activation of a pentafluorophenylethylamine derivative: Formation of glutathione conjugates in vitro in the rat

J. Shang; Shiyao Xu; Yohannes Teffera; George A. Doss; Ralph A. Stearns; S. Edmonson; Maria Beconi

The aim was to investigate the metabolic activation potential of a pentafluorophenylethylamine derivative (compound I) in vitro in the rat and to identify the cytochrome P450 (CYP) enzymes that catalyse these metabolic activation processes. Reduced glutathione (GSH) was fortified in rat hepatocytes and liver microsomes to trap possible reactive intermediates. Four glutathione conjugates (M1–4) were identified by LC-MSn following incubation of compound I in GSH-enriched rat hepatocytes and liver microsomes. Three of these conjugates (M2–4) have not been reported previously for pentafluorophenyl derivatives. Elemental composition analysis of these conjugates was obtained using high-resolution quadrupole time-of-flight mass spectrometry. The formation of GSH conjugate M1 was rationalized as a direct nucleophilic replacement of fluoride by glutathione, whereas the formation of the GSH conjugates M2–4 was proposed to occur by NADPH-dependent metabolic activation of the pentafluorophenyl ring via arene oxide, quinone and/or quinoneimine reactive intermediates. Formation of these conjugates was enhanced three- to five-fold in liver microsomes obtained from phenobarbital- and dexamethasone-treated rats. In incubations with pooled rat liver microsomes and recombinant rat CYP3A1 and CYP3A2, troleandomycin (TAO) reduced the formation of GSH conjugates M2–4 by 80–90%, but it had no effect on the formation of M1. Incubation of compound I with rat supersomes indicated that only CYP3A1 and CYP3A2 were capable of mediating these metabolic activation processes.


PLOS ONE | 2018

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

Daniel Elbaum; Maria Beconi; Edith Monteagudo; Annalise Di Marco; Maria S. Quinton; Kathryn A. Lyons; Andrew Vaino; Steven Harper

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.


Journal of Chromatography B | 2018

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

Roberto Speziale; Camilla Montesano; Maria Lucia de Leonibus; Fabio Bonelli; Paola Fezzardi; Maria Beconi; Edith Monteagudo; Daniel Elbaum; Laura Orsatti

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.

Collaboration


Dive into the Maria Beconi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Wityak

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge