12 results on '"Elizabeth M. Joshi"'
Search Results
2. Informing the Selection of Screening Hit Series with in Silico Absorption, Distribution, Metabolism, Excretion, and Toxicity Profiles
- Author
-
John M, Sanders, Douglas C, Beshore, J Christopher, Culberson, James I, Fells, Jason E, Imbriglio, Hakan, Gunaydin, Andrew M, Haidle, Marc, Labroli, Brian E, Mattioni, Nunzio, Sciammetta, William D, Shipe, Robert P, Sheridan, Linda M, Suen, Andreas, Verras, Abbas, Walji, Elizabeth M, Joshi, and Tjerk, Bueters
- Subjects
Pharmacology ,Drug-Related Side Effects and Adverse Reactions ,Pharmaceutical Preparations ,Drug Discovery ,Animals ,Humans ,Quantitative Structure-Activity Relationship ,Computer Simulation ,Pharmacokinetics ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,High-Throughput Screening Assays - Abstract
High-throughput screening (HTS) has enabled millions of compounds to be assessed for biological activity, but challenges remain in the prioritization of hit series. While biological, absorption, distribution, metabolism, excretion, and toxicity (ADMET), purity, and structural data are routinely used to select chemical matter for further follow-up, the scarcity of historical ADMET data for screening hits limits our understanding of early hit compounds. Herein, we describe a process that utilizes a battery of in-house quantitative structure-activity relationship (QSAR) models to generate in silico ADMET profiles for hit series to enable more complete characterizations of HTS chemical matter. These profiles allow teams to quickly assess hit series for desirable ADMET properties or suspected liabilities that may require significant optimization. Accordingly, these in silico data can direct ADMET experimentation and profoundly impact the progression of hit series. Several prospective examples are presented to substantiate the value of this approach.
- Published
- 2017
3. Discovery of a Potent, Dual Serotonin and Norepinephrine Reuptake Inhibitor
- Author
-
Gustavo Pascual, Smriti Iyengar, Gregory A. Stephenson, Laura J. Martin, Sandra Ann Filla, Craig E. Thomas, Susan K. Hemrick-Luecke, Kirk W. Johnson, Jikesh Arvind Shah, Eric George Tromiczak, Magnus Wilhelm Walter, Karsten Fynboe, Lance Allen Pfeifer, Mark A. Muhlhauser, Juan A. Rincón, Nicolas Dreyfus, Chunjin Ding, Jia Shaojuan, María Luz de la Puente, Beverly A. Heinz, Lee A. Phebus, Michael P. Johnson, Simon James Richards, Thomas J. Perun, Michael J. McCoy, Pilar Lopez, Hamideh Zarrinmayeh, Thierry Masquelin, Rosa Maria A. Simmons, Douglas Linn Gernert, Oscar de Frutos, Elizabeth M. Joshi, Denise Morrow, Wayne W. Weber, Anette M. Johansson, Linda K. Thompson, Eric P. Seest, Valentina O. Badescu, Javier Mendiola, Jason K. Myers, and Patrick L. Love
- Subjects
biology ,business.industry ,Organic Chemistry ,Chronic pain ,Pharmacology ,Inhibitory postsynaptic potential ,medicine.disease ,Biochemistry ,Norepinephrine reuptake inhibitor ,Norepinephrine transporter ,In vivo ,Drug Discovery ,medicine ,biology.protein ,Serotonin ,Reuptake inhibitor ,business ,ADME - Abstract
The objective of the described research effort was to identify a novel serotonin and norepinephrine reuptake inhibitor (SNRI) with improved norepinephrine transporter activity and acceptable metabolic stability and exhibiting minimal drug-drug interaction. We describe herein the discovery of a series of 3-substituted pyrrolidines, exemplified by compound 1. Compound 1 is a selective SNRI in vitro and in vivo, has favorable ADME properties, and retains inhibitory activity in the formalin model of pain behavior. Compound 1 thus represents a potential new probe to explore utility of SNRIs in central nervous system disorders, including chronic pain conditions.
- Published
- 2013
- Full Text
- View/download PDF
4. Development of LC-MS/MS-Based Receptor Occupancy Tracers and Positron Emission Tomography Radioligands for the Nociceptin/Orphanin FQ (NOP) Receptor
- Author
-
Michael A. Statnick, Karen S. Rash, Concepción Pedregal, Steven D. Kahl, Daniel R. Mudra, Zhaogen Chen, Miguel A. Toledo, Antonio Navarro, Ana Benito, Nuria Diaz, Elizabeth M. Joshi, Vanessa N. Barth, Celia Lafuente, Alma Jiménez, and Maria Angeles Martinez-Grau
- Subjects
Male ,Narcotic Antagonists ,NOP ,CHO Cells ,Thiophenes ,Pharmacology ,Nociceptin Receptor ,Rats, Sprague-Dawley ,Radioligand Assay ,Structure-Activity Relationship ,Cricetulus ,Tandem Mass Spectrometry ,In vivo ,Cricetinae ,Drug Discovery ,Radioligand ,medicine ,Animals ,Humans ,Structure–activity relationship ,Spiro Compounds ,Carbon Radioisotopes ,Receptor ,medicine.diagnostic_test ,Chemistry ,Brain ,Stereoisomerism ,Rats ,Nociceptin receptor ,HEK293 Cells ,Positron emission tomography ,Positron-Emission Tomography ,Receptors, Opioid ,Macaca ,Molecular Medicine ,Radiopharmaceuticals ,Chromatography, Liquid - Abstract
Currently, a lack of sufficient tools has limited the understanding of the relationship between neuropsychiatric disorders and the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor. Herein, we describe the discovery and development of an antagonist NOP receptor occupancy (RO) tracer and a novel positron emission tomography (PET) radioligand suitable to probe the NOP receptor in human clinical studies. A thorough structure-activity relationship (SAR) around the high-affinity 3-(2'-fluoro-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]-1-yl)-2-(2-halobenzyl)-N-alkylpropanamide scaffold identified a series of subnanomolar, highly selective NOP antagonists. Subsequently, these unlabeled NOP ligands were evaluated in vivo by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in rat to determine brain uptake, kinetics and specific binding. (S)-27 was identified as a suitable unlabeled preclinical RO tracer to accurately quantify NOP receptor engagement in rat brain. Three compounds were selected for evaluation in nonhuman primates as PET tracers: (-)-26, (-)-30, and (-)-33. Carbon-11 labeling of (+)-31 yielded [(11)C]-(S)-30, which exhibited minimal generation of central nervous system (CNS) penetrant radiometabolites, improved brain uptake, and was an excellent PET radioligand in both rat and monkey. Currently [(11)C]-(S)-30 is being evaluated as a PET radiotracer for the NOP receptor in human subjects.
- Published
- 2012
- Full Text
- View/download PDF
5. Target Engagement for PK/PD Modeling and Translational Imaging Biomarkers
- Author
-
Elizabeth M. Joshi, Matthew D. Silva, and Vanessa N. Barth
- Subjects
Target engagement ,Computational biology ,Biology ,Bioinformatics ,PK/PD models - Published
- 2012
- Full Text
- View/download PDF
6. 2-ABT-S-oxide detoxification by glutathioneS-transferases A1-1, M1-1 and P1-1: Implications for toxicity associated with zileuton
- Author
-
Timothy L. Macdonald, Elizabeth M. Joshi, and Brian H. Heasley
- Subjects
Magnetic Resonance Spectroscopy ,Health, Toxicology and Mutagenesis ,Thiophenes ,Urine ,Pharmacology ,Toxicology ,Biochemistry ,Mass Spectrometry ,chemistry.chemical_compound ,Detoxification ,Toxicity Tests ,medicine ,Hydroxyurea ,Glutathione Transferase ,Leukotriene ,Molecular Structure ,biology ,General Medicine ,Glutathione ,Zileuton ,In vitro ,Isoenzymes ,Kinetics ,Glutathione S-transferase ,Liver ,chemistry ,Inactivation, Metabolic ,Toxicity ,biology.protein ,Chromatography, Liquid ,medicine.drug - Abstract
Zileuton, an agent which targets the leukotriene pathway through inhibition of 5-lipoxygenase (5-LO), was approved for the treatment of asthma in 1997. Shortly after its release, its use was restricted due to the observation of hepatotoxicity in patients. Previous research from the authors' laboratory demonstrated the formation of the reactive metabolite, 2-ABT-S-oxide (M1) from zileuton, and has identified a mercapturate of 2-ABT, C1, in the urine of rats dosed with zileuton. The reaction between M1 and glutathione (GSH) has been established in vitro; however, the potential for catalysis by glutathione transferases (GSTs) was not addressed. The work presented here outlines a role for GSTs in the detoxification of M1. Non-enzymatic conjugation studies with M1 and GSH in control experiments led to a t(1/2) of 6.4 +/- 0.4 h at pH 6.5. This rate was accelerated in the presence of GSTA1-1, GSTM1-1 and GSTP1-1 providing t(1/2) values of 2.6 +/- 0.1, 0.53 +/- 0.02, and 0.3 +/- 0.04 h, respectively, at pH 6.5. The inhibition of various GST enzymes was also studied. Results show that M1 inhibits GSTM1-1 and GSTP1-1 to a greater extent as compared with GSTA1-1. In the case of GSTA1-1, the inhibition was observed to be reversible, whereas M1 inhibition of GSTM1-1 and GSTP1-1 was found to be irreversible under identical conditions. GSTM1-1 is present in liver and thus the finding of the alkylation and potential irreversible inactivation of this isoform in vivo could contribute to an understanding of the hepatotoxicity associated with zileuton.
- Published
- 2009
- Full Text
- View/download PDF
7. Synthesis of diketopiperazine-based carboline homodimers and in vitro growth inhibition of human carcinomas
- Author
-
Amy M. Deveau, Timothy L. Macdonald, Elizabeth M. Joshi, and Nancy E. Costa
- Subjects
Models, Molecular ,Stereochemistry ,Dimer ,Clinical Biochemistry ,Pharmaceutical Science ,Diketopiperazines ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cell Line, Tumor ,Drug Discovery ,Humans ,Cytotoxicity ,Molecular Biology ,Cell Proliferation ,Indole test ,Molecular Structure ,Carcinoma ,Organic Chemistry ,Tryptophan ,Stereoisomerism ,chemistry ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Enantiomer ,Growth inhibition ,Dimerization ,Lead compound ,Carbolines - Abstract
Starting from d - or l -tryptophan, we have synthesized and characterized six compounds 2.29 – 2.31a and b that belong to a class of nitrogen heterocycles: the carboline-based homodimers. Each individual homodimer features a 1,3- trans relationship on each side of the central diketopiperazine core, but differs in absolute stereochemistry and also in substitution on the 4′ and 4″ oxygens (–Bn, –CH 3 , or –H). The in vitro cytotoxicity of the six compounds was evaluated by measuring the growth inhibition in NCI–H520 and PC-3 human carcinoma cells. Phenol 2.30a inhibited cancer cell growth approximately three times better than its enantiomer 2.30b and possessed a GI 50 comparable to the clinically used agent etoposide in both cell lines. We have concluded that both the stereochemistry imparted by l -tryptophan and the presence of hydroxy substituents at the 4′ and 4″ positions are necessary to generate cytotoxic properties in the homodimer class. We are now employing 2.30a as a new lead compound in our efforts to discover improved indole-based cancer chemotherapeutics.
- Published
- 2008
- Full Text
- View/download PDF
8. Regioselective oxidation of 2-amino-3-aroyl-4,5-dialkylthiophenes by DMSO
- Author
-
Timothy L. Macdonald, Elizabeth M. Joshi, Joel Linden, Ray A. Olsson, and Mahendra D. Chordia
- Subjects
Reaction mechanism ,Ketone ,Clinical Biochemistry ,Pharmaceutical Science ,chemistry.chemical_element ,Thiophenes ,Biochemistry ,Medicinal chemistry ,chemistry.chemical_compound ,Drug Discovery ,Organic chemistry ,Dimethyl Sulfoxide ,Molecular Biology ,chemistry.chemical_classification ,Molecular Structure ,Dimethyl sulfoxide ,organic chemicals ,Organic Chemistry ,Regioselectivity ,Benzothiophene ,General Medicine ,Thiophene derivatives ,chemistry ,Molecular Medicine ,Oxidation-Reduction ,Carbon - Abstract
Solutions of 2-amino-3-aroyl-4,5-dialkylthiophenes in DMSO (dimethyl sulfoxide) undergo regioselective oxidation of benzylic carbon under mild conditions. We describe three examples and propose a mechanism for oxidation.
- Published
- 2004
- Full Text
- View/download PDF
9. In Vitro Metabolism of 2-Acetylbenzothiophene: Relevance to Zileuton Hepatotoxicity
- Author
-
Elizabeth M. Joshi, Timothy L. Macdonald, Brian H. Heasley, and Mahendra D. Chordia
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Magnetic Resonance Spectroscopy ,Metabolite ,Thiophenes ,Toxicology ,Cell Line ,Adduct ,chemistry.chemical_compound ,In vivo ,medicine ,Animals ,Humans ,Hydroxyurea ,Lymphoblast ,General Medicine ,Zileuton ,Glutathione ,In vitro ,Rats ,Liver ,Biochemistry ,chemistry ,Microsome ,Half-Life ,medicine.drug - Abstract
Zileuton, an inhibitor of 5-lipooxygenase, the initial enzyme in the leukotriene pathway, was marketed as a new treatment for asthma. This drug has been associated with liver toxicity, which has limited its clinical usefulness. We provide evidence here that the liver toxicity likely involves a sequence of biotransformations leading to 2-acetylbenzothiophene (2-ABT), which is subsequently metabolized to give a reactive intermediate(s). In vitro experiments with the human lymphoblast MCL5 cell line demonstrated that 2-ABT is cytotoxic in a P450-dependent manner. Human liver microsome (HLM) incubations with 2-ABT revealed the formation of two short-lived oxidized species, "M + 16" and "M + 32". Both of these metabolites formed adducts in the presence of GSH or NAC. Singly oxidized M + 16 adducts, from either GSH or NAC, appeared to be unstable in acidic medium and eliminated water readily to form a new compound. Authentic synthetic standards demonstrated that 2-ABT-S-oxide M1 corresponded to the M + 16 metabolite and that the S-oxide underwent nucleophilic addition with GSH and NAC to produce the singly oxidized adducts observed in HLM. The S-oxide adducts readily eliminated water to form a rearomatized 2-ABT-GSH adduct or 2-ABT-NAC adduct. Coelution experiments with the synthetic standard confirmed the structure of the eliminated 2-ABT-NAC adduct C1. LC/MS analyses of urine samples collected from rats dosed with zileuton indicate that C1 is a metabolite of zileuton formed in vivo. The in vitro and in vivo data presented here demonstrate the formation of 2-ABT from zileuton and its further bioactivation to a potentially toxic metabolite.
- Published
- 2004
- Full Text
- View/download PDF
10. Efficiency gains in tracer identification for nuclear imaging: can in vivo LC-MS/MS evaluation of small molecules screen for successful PET tracers?
- Author
-
Dana Rae Benesh, Lee A. Phebus, Zhaogen Chen, Charles H. Mitch, Anne B. Need, John Mehnert Schaus, Elizabeth M. Joshi, Thomas J. Raub, Vanessa N. Barth, and Stuart Morton
- Subjects
Physiology ,Cognitive Neuroscience ,Standardized uptake value ,Mass spectrometry ,Biochemistry ,Mice ,In vivo ,Tandem Mass Spectrometry ,TRACER ,medicine ,Animals ,Humans ,Pet tracer ,medicine.diagnostic_test ,Chemistry ,business.industry ,Binding potential ,Brain ,Cell Biology ,General Medicine ,Small molecule ,Rats ,Positron emission tomography ,Positron-Emission Tomography ,Biophysics ,Radiopharmaceuticals ,Nuclear medicine ,business ,Biomarkers ,Chromatography, Liquid - Abstract
Positron emission tomography (PET) imaging has become a useful noninvasive technique to explore molecular biology within living systems; however, the utility of this method is limited by the availability of suitable radiotracers to probe specific targets and disease biology. Methods to identify potential areas of improvement in the ability to predict small molecule performance as tracers prior to radiolabeling would speed the discovery of novel tracers. In this retrospective analysis, we characterized the brain penetration or peak SUV (standardized uptake value), binding potential (BP), and brain exposure kinetics across a series of known, nonradiolabeled PET ligands using in vivo LC-MS/MS (liquid chromatography coupled to mass spectrometry) and correlated these parameters with the reported PET ligand performance in nonhuman primates and humans available in the literature. The PET tracers studied included those reported to label G protein-coupled receptors (GPCRs), intracellular enzymes, and transporters. Additionally, data for each tracer was obtained from a mouse brain uptake assay (MBUA), previously published, where blood-brain barrier (BBB) penetration and clearance parameters were assessed and compared against similar data collected on a broad compound set of central nervous system (CNS) therapeutic compounds. The BP and SUV identified via nonradiolabeled LC-MS/MS, while different from the published values observed in the literature PET tracer data, allowed for an identification of initial criteria values we sought to facilitate increased potential for success from our early discovery screening paradigm. Our analysis showed that successful, as well as novel, clinical PET tracers exhibited BP of greater than 1.5 and peak SUVs greater than approximately 150% at 5 min post dose in rodents. The brain kinetics appeared similar between both techniques despite differences in tracer dose, suggesting linearity across these dose ranges. The assessment of tracers in a CNS exposure model, the mouse brain uptake assessment (MBUA), showed that those compound with initial brain-to-plasma ratios2 and unbound fraction in brain homogenate0.01 were more likely to be clinically successful PET ligands. Taken together, early incorporation of a LC/MS/MS cold tracer discovery assay and a parallel MBUA can be an useful screening paradigm to prioritize and rank order potential novel PET radioligands during early tracer discovery efforts. Compounds considered for continued in vivo PET assessments can be identified quickly by leveraging in vitro affinity and selectivity measures, coupled with data from a MBUA, primarily the 5 min brain-to-plasma ratio and unbound fraction data. Coupled utilization of these data creates a strategy to efficiently screen for the identification of appropriate chemical space to invest in for radiotracer discovery.
- Published
- 2014
11. Evaluation in vitro and in animals of a new 11C-labeled PET radioligand for metabotropic glutamate receptors 1 in brain
- Author
-
John Joseph Masters, Robert B. Innis, Johannes Tauscher, Victor W. Pike, Paolo Zanotti-Fregonara, David T. Clark, Bruce A. Dressman, Emily Rhoads, Debra Luffer-Atlas, Sami S. Zoghbi, Vanessa N. Barth, Nancy Goebl, Beverly A. Heinz, Cheryl L. Morse, Kuklish Steven Lee, Elizabeth M. Joshi, Edward R. Siuda, Jeih San Liow, Matthew J. Fisher, and Eric S. Nisenbaum
- Subjects
Male ,Spectrometry, Mass, Electrospray Ionization ,Allosteric regulation ,Pharmacology ,In Vitro Techniques ,Ligands ,Receptors, Metabotropic Glutamate ,Article ,Inhibitory Concentration 50 ,Mice ,Tandem Mass Spectrometry ,mental disorders ,Radioligand ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Mice, Knockout ,Carbon Isotopes ,medicine.diagnostic_test ,Metabotropic glutamate receptor 5 ,Chemistry ,business.industry ,Brain ,General Medicine ,In vitro ,Rats ,Models, Chemical ,Metabotropic glutamate receptor ,Positron emission tomography ,Blood-Brain Barrier ,Positron-Emission Tomography ,Metabotropic glutamate receptor 1 ,Nuclear medicine ,business ,Allosteric Site ,Chromatography, Liquid - Abstract
Two allosteric modulators of the group I metabotropic glutamate receptors (mGluR1 and mGluR5) were evaluated as positron emission tomography (PET) radioligands for mGluR1.LY2428703, a full mGluR1 antagonist (IC(50) 8.9 nM) and partial mGluR5 antagonist (IC(50) 118 nM), and LSN2606428, a full mGluR1 and mGluR5 antagonist (IC(50) 35.3 nM and 10.2 nM, respectively) were successfully labeled with (11)C and evaluated as radioligands for mGluR1. The pharmacology of LY2428703 was comprehensively assessed in vitro and in vivo, and its biodistribution was investigated by liquid chromatography-mass spectrometry/mass spectrometry, and by PET imaging in the rat. In contrast, LSN2606428 was only evaluated in vitro; further evaluation was stopped due to its unfavorable pharmacological properties and binding affinity.(11)C-LY2428703 showed promising characteristics, including: (1) high potency for binding to human mGluR1 (IC(50) 8.9 nM) with no significant affinity for other human mGlu receptors (mGluR2 through mGluR8); (2) binding to brain displaceable by administration of an mGluR1 antagonist; (3) only one major radiometabolite in both plasma and brain, with a negligible brain concentration (with 3.5 % of the total radioactivity in cerebellum) and no receptor affinity; (4) a large specific and displaceable signal in the mGluR1-rich cerebellum with no significant in vivo affinity for mGluR5, as shown by PET studies in rats; and (5) lack of substrate behavior for efflux transporters at the blood-brain barrier, as shown by PET studies conducted in wild-type and knockout mice.(11)C-LY2428703, a new PET radioligand for mGluR1 quantification, displayed promising characteristics both in vitro and in vivo in rodents.
- Published
- 2012
12. ChemInform Abstract: Synthesis of Diketopiperazine-Based Carboline Homodimers and in vitro Growth Inhibition of Human Carcinomas
- Author
-
Timothy L. Macdonald, Elizabeth M. Joshi, Nancy E. Costa, and Amy M. Deveau
- Subjects
Indole test ,chemistry.chemical_compound ,chemistry ,Stereochemistry ,Cell culture ,Cancer cell ,Tryptophan ,Cytotoxic T cell ,General Medicine ,Enantiomer ,Growth inhibition ,Lead compound - Abstract
Starting from d - or l -tryptophan, we have synthesized and characterized six compounds 2.29 – 2.31a and b that belong to a class of nitrogen heterocycles: the carboline-based homodimers. Each individual homodimer features a 1,3- trans relationship on each side of the central diketopiperazine core, but differs in absolute stereochemistry and also in substitution on the 4′ and 4″ oxygens (–Bn, –CH 3 , or –H). The in vitro cytotoxicity of the six compounds was evaluated by measuring the growth inhibition in NCI–H520 and PC-3 human carcinoma cells. Phenol 2.30a inhibited cancer cell growth approximately three times better than its enantiomer 2.30b and possessed a GI 50 comparable to the clinically used agent etoposide in both cell lines. We have concluded that both the stereochemistry imparted by l -tryptophan and the presence of hydroxy substituents at the 4′ and 4″ positions are necessary to generate cytotoxic properties in the homodimer class. We are now employing 2.30a as a new lead compound in our efforts to discover improved indole-based cancer chemotherapeutics.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.