A Two-Point IC50 Method for Evaluating the Biochemical Potency of Irreversible Enzyme Inhibitors

Total Page:16

File Type:pdf, Size:1020Kb

A Two-Point IC50 Method for Evaluating the Biochemical Potency of Irreversible Enzyme Inhibitors bioRxiv preprint doi: https://doi.org/10.1101/2020.06.25.171207; this version posted June 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. A two-point IC50 method for evaluating the biochemical potency of irreversible enzyme inhibitors Petr Kuzmicˇ BioKin Ltd., Watertown, Massachusetts, USA http://www.biokin.com Abstract Irreversible (covalent) enzyme inhibitors cannot be unambiguously ranked for biochemical potency by using IC50 values determined at a single point in time, because the same IC50 value could originate either from relatively low initial binding affinity accompanied by high chemical reactivity, or the other way around. To disambiguate the potency ranking of covalent inhibitors, here we describe a data-analytic procedure relying on two separate IC50 values, determined at two different reaction times. In the case of covalent inhibitors following the two-step kinetic mechanism E + I E·I ! EI, the two IC50 values alone can be used to estimate both the inhibition constant (Ki) as a measure of binding affinity and the inactivation rate constant (kinact) as a measure of chemical reactivity. In the case of covalent inhibitors following the one-step kinetic mechanism E + I ! EI, a simple algebraic formula can be used to estimate the covalent efficiency constant (kinact=Ki) from a single experimental value of IC50. The two simplifying assumptions underlying the method are (1) zero inhibitor depletion, which implies that the inhibitor concentrations are always significantly higher than the enzyme concentration; and (2) constant reaction rate in the uninhibited control assay. The newly proposed method is validated by using a simulation study involving 64 irreversible inhibitors with covalent efficiency constants spanning seven orders of magnitude. Key words: enzyme kinetics; inhibition; irreversible inhibition; covalent inhibition; mathematical model; algebraic model 1. Introduction reporting the I50 for an irreversible inhibitor without also spec- ifying the corresponding reaction time is essentially meaning- Many medicines currently in use to treat various human dis- less. An even more serious conceptual problem is that the same eases and symptoms are enzyme inhibitors. Furthermore, many value of I50 could originate either from relatively high initial important drugs and drug candidates are irreversible covalent binding affinity (low Ki) and relatively low chemical reactiv- inhibitors [1–4], which express their pharmacological effect by ity (low kinact), or the other way around. Therefore, two or more forming a permanent chemical bond with the protein target. inhibitors with disparate molecular properties could easily man- Probably the most well known representative of this class is ifest as having “the same” biochemical potency if the I50 assay acetylsalicylate, or Aspirin, an irreversible covalent inhibitor of is conducted at a single point in time. cyclooxygenase. Evaluating the biochemical potency of irre- This report presents a data-analytic procedure that relies on versible inhibitors in the process of pre-clinical drug discovery two separate I50 determinations, conducted at two different re- is exceedingly challenging. Even the task of simply arranging action times. If a given inhibitor follows a stepwise mecha- a list of potential drug candidates in order of their biochemi- nism of inhibition, involving a kinetically detectable noncova- cal potency presents a serious obstacle. The main challenge is lent intermediate, we show that it is possible to estimate both that the overall biochemical potency of irreversible enzyme in- Ki and kinact from the two time-dependent I50 measurements hibitors has two distinct components, namely, binding affinity alone. Many highly potent covalent inhibitors display a one- measured by the inhibition constant (Ki) and chemical reactiv- step kinetic mechanism, apparently without the involvement of ity measured by the inactivation rate constant (kinact). a clearly detectable noncovalent intermediate [6]. In those cases Medicinal chemists and pharmacologists involved in drug it is in principle impossible to measure the Ki and kinact sepa- discovery are accustomed to expressing the biochemical po- rately, but the data-analytic method described here allows for tency of enzyme inhibitors primarily in terms of the I [5]1. 50 the determination of the covalent efficiency constant keff, also However, in the case of irreversible inhibitors, the I by defi- 50 known as kinact=Ki, from any single measurement of I50. The nition decreases over time, until at asymptotically infinite time proposed method is validated by using a simulation study in- it approaches one half of the enzyme concentration. Therefore, volving 64 computer-generated inhibitors with molecular prop- erties (Ki, kinact, and kinact=Ki) spanning at least six orders of 1 magnitude. Throughout this manuscript, the conventionally used notation IC50 is ab- breviated as I50. BioKin Technical Note TN-2020-03 Revision 1.14 :: 24 June 2020 bioRxiv preprint doi: https://doi.org/10.1101/2020.06.25.171207; this version posted June 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 2. Methods 2.2.2. Model discrimination analysis On the assumption that the enzyme assay proceeds kinet- This section describes the theoretical and mathematical meth- ically via the one-step inhibition mechanism C, the apparent ods that were used in heuristic simulations described in this re- ∗ second-order rate constant k1 is by definition invariant with re- port. All computations were performed by using the software spect to time, according to Eqn (1). Thus, in the idealized case package DynaFit [7, 8]. ∗(1) ∗(2) of zero experimental error, two values k1 and k1 determined at two different stopping times t(1) and t(2) should be exactly 2.1. Kinetic mechanisms of irreversible inhibition identical. However, in the realistic case of non-zero experi- In this report we will consider in various contexts the kinet- ∗ mental error, the two k1 values will always be ever so slightly ics mechanisms of substrate catalysis and irreversible inhibition different even if the one-step kinetic mechanism C is actually depicted in Figure 1. For further details see ref. [9]. ∗(1) ∗(2) operating. In order to decide whether or not k1 and k1 are sufficiently similar to warrant the acceptance of the one-step k1s k2s kinetic model, we use the geometric standard deviation defined E + S E•S E + P by Eqn (4), where µg is the geometric mean defined by Eqn (3). k-1s q µ = ∗(1) ∗(2) g k1 k1 (3) k1 k2 "A": E + I E•I EI tuv 0 ∗ 12 0 ∗ 12 k-1 B (1) C B (1) C B k1 C B k1 C σg = exp @Bln AC + @Bln AC (4) µg µg Ki k2 "B": E + I E•I EI The maximum acceptable value of σg will depend on the Ki = k-1 / k1 experimental situation. In the simulation study reported below, where the pseudo-random noise was equal to one percent of the σ < k1 maximum signal (e.g., fluorescence value), we found that g "C": E + I EI 1:25 was producing satisfactory results. Note that the geometric standard deviation σg is a dimensionless quantity measuring the Figure 1: Kinetic mechanisms of substrate catalysis (top) and “X-fold variation” associated with a group of numerical values. covalent inhibition (mechanisms A – C). σ < : ∗(1) In our situation, g 1 25 means that the two values k1 and ∗(2) k1 in a statistical sense differ by less than a factor of 1.25, or roughly by 25 percent in either direction (lower or higher). 2.2. Mathematical models ∗ ∗ 2.2.3. Determination of kinact and K from two values of I50 2.2.1. Determination of k1 from a single measurement of I50 i On the assumption that the enzyme assay proceeds kinet- On the assumption that the enzyme assay proceeds kinet- ically via the one-step inhibition mechanism C, the apparent ically via the two-step inhibition mechanism B, the apparent ∗ ∗ = = + = inhibition constant K = Ki (1 + [S]0=KM) can be computed second-order rate constant k1 k1 (1 [S]0 KM) can be com- i directly from any two measurements of I by using Eqn (5), puted directly from a single measurement of I50 by using Eqn 50 where t(1) < t(2) are the two reaction times used to determine (1), where t50 is the reaction time used to determine the ex- 50 50 (1) (2) perimental value of I50 and c is a constant depending on the I50 and I50 , respectively, and a is an empirical constant (see dimensions used to express time and concentration. When time below). is expressed in seconds and concentrations in micromoles per ( ) liter, c = 1:5936. The requisite algebraic derivation is shown in 1=a 1 − t(1)=t(2) the Supporting Information. Eqn (2) defines the second-order ∗ = 50 50 Ki ( ) = (5) covalent efficiency constant in the context of the one-step mech- (1) (2) 1 a 1 t =t anism C. − 50 50 (1) (2) I50 I50 2 0 1 3 c ∗ k∗ = (1) 1 6 B K C 7 1 k = 6a B i − C + b7 I50 t50 inact (2) exp 4 ln @ (2) 1A 5 (6) ! t50 I50 ∗ [S]0 ! k ff = k 1 + (2) e 1 K kinact [S]0 M keff = ∗ 1 + (7) Ki KM ∗ Once Ki is determined from Eqn (5), kinact can be computed (2) from it and from I50 by using Eqn (6), where a and b are em- 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.06.25.171207; this version posted June 27, 2020.
Recommended publications
  • Hypochlorous Acid Handling
    Hypochlorous Acid Handling 1 Identification of Petitioned Substance 2 Chemical Names: Hypochlorous acid, CAS Numbers: 7790-92-3 3 hypochloric(I) acid, chloranol, 4 hydroxidochlorine 10 Other Codes: European Community 11 Number-22757, IUPAC-Hypochlorous acid 5 Other Name: Hydrogen hypochlorite, 6 Chlorine hydroxide List other codes: PubChem CID 24341 7 Trade Names: Bleach, Sodium hypochlorite, InChI Key: QWPPOHNGKGFGJK- 8 Calcium hypochlorite, Sterilox, hypochlorite, UHFFFAOYSA-N 9 NVC-10 UNII: 712K4CDC10 12 Summary of Petitioned Use 13 A petition has been received from a stakeholder requesting that hypochlorous acid (also referred 14 to as electrolyzed water (EW)) be added to the list of synthetic substances allowed for use in 15 organic production and handling (7 CFR §§ 205.600-606). Specifically, the petition concerns the 16 formation of hypochlorous acid at the anode of an electrolysis apparatus designed for its 17 production from a brine solution. This active ingredient is aqueous hypochlorous acid which acts 18 as an oxidizing agent. The petitioner plans use hypochlorous acid as a sanitizer and antimicrobial 19 agent for the production and handling of organic products. The petition also requests to resolve a 20 difference in interpretation of allowed substances for chlorine materials on the National List of 21 Allowed and Prohibited Substances that contain the active ingredient hypochlorous acid (NOP- 22 PM 14-3 Electrolyzed water). 23 The NOP has issued NOP 5026 “Guidance, the use of Chlorine Materials in Organic Production 24 and Handling.” This guidance document clarifies the use of chlorine materials in organic 25 production and handling to align the National List with the November, 1995 NOSB 26 recommendation on chlorine materials which read: 27 “Allowed for disinfecting and sanitizing food contact surfaces.
    [Show full text]
  • Exploring the Concept of Safety and Tolerability
    SECOND ANNUAL WORKSHOP ON CLINICAL OUTCOME ASSESSMENTS IN CANCER CLINICAL TRIALS April 25, 2017 Bethesda, MD Co-sponsored by Session 1 Exploring the Concepts of Safety and Tolerability: Incorporating the Patient Voice SECOND ANNUAL WORKSHOP ON CLINICAL OUTCOME ASSESSMENTS IN CANCER CLINICAL TRIALS April 25, 2017 Bethesda, MD Co-sponsored by Disclaimer • The views and opinions expressed in the following slides are those of the individual presenters and should not be attributed to their respective organizations/companies, the U.S. Food and Drug Administration or the Critical Path Institute. • These slides are the intellectual property of the individual presenters and are protected under the copyright laws of the United States of America and other countries. Used by permission. All rights reserved. All trademarks are the property of their respective owners. 3 Session Participants Chair • Bindu Kanapuru, MD – Medical Officer, Division of Hematology Products, OHOP, FDA Presenters • James (Randy) Hillard, MD – Professor of Psychiatry, Michigan State University • Crystal Denlinger, MD, FACP – Associate Professor, Department of Hematology/Oncology; Chief, Gastrointestinal Medical Oncology; Director, Survivorship Program; Deputy Director, Phase 1 Program, Fox Chase Cancer Center • Katherine Soltys, MD – Acting Director, Bureau of Medical Sciences, Therapeutic Products Directorate, Health Products and Food Branch, Health Canada • Karen E. Arscott, DO, MSc – Associate Professor of Medicine-Patient Advocate and Survivor, Geisinger Commonwealth
    [Show full text]
  • Bugs, Drugs, and Cancer: Can the Microbiome Be a Potential
    REVIEWS Drug Discovery Today Volume 24, Number 4 April 2019 Reviews Bugs, drugs, and cancer: can the GENE TO SCREEN microbiome be a potential therapeutic target for cancer management? 1,z 2,z 1 1 Biying Chen , Guangye Du , Jiahui Guo and Yanjie Zhang 1 Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201999, China 2 Department of Pathology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201999, China Outnumbering our own cells over ten times, gut microbes can even be considered an additional organ. Several studies have explored the association between microbiomes and antitumor drug response. It has been reported that the presence of specific bacteria might modulate cancer progression and the efficacy of anticancer therapeutics. Bacteria-targeting intervention can provide crucial guidance for the design of next-generation antitumor drugs. Here, we review previous findings elucidating the impact of gut microbiomes on cancer treatment and the possible underlying mechanisms. In addition, we examine the role of microbiome manipulation in controlling tumor growth. Finally, we discuss concerns regarding the alteration of the microbiome composition, and the potential approaches to surpass existing limitations. Introduction cantly during life [5] (Fig. 1). Gut microbial density increases from The microbiota is defined as all microorganisms that are associated the proximal to the distal GI tract and along the tissue–lumen axis. with a specific host cellular environment [1]. These microorganisms Similarly, diversity further increases in the same pattern [6]. More- are identified using 16S ribosomal RNA (rRNA)sequencing.
    [Show full text]
  • Assessment of Combined Toxicity of Heavy Metals from Industrial Wastewaters on Photobacterium Phosphoreum T3S
    Appl Water Sci DOI 10.1007/s13201-016-0385-4 ORIGINAL ARTICLE Assessment of combined toxicity of heavy metals from industrial wastewaters on Photobacterium phosphoreum T3S 1,2 1 2 1 2 BibiSaima Zeb • Zheng Ping • Qaisar Mahmood • Qiu Lin • Arshid Pervez • 2 2 2 Muhammad Irshad • Muhammad Bilal • Zulfiqar Ahmad Bhatti • Shahida Shaheen2 Received: 19 May 2015 / Accepted: 19 January 2016 Ó The Author(s) 2016. This article is published with open access at Springerlink.com Abstract This research work is focusing on the toxicities Introduction of heavy metals of industrial origin to anaerobic digestion of the industrial wastewater. Photobacterium phospho- The presence of heavy metals in excess amounts inter- reum T3S was used as an indicator organism. The acute feres with the beneficial uses of water because of the toxicities of heavy metals on P. phosphoreum T3S were toxicity of heavy metals and the biomagnification effect assessed during 15-min half inhibitory concentration brought on by its accumulation on ecology (Chang et al. (IC50) as indicator at pH 5.5–6. Toxicity assays involved 2006; Altas 2009). During recent times, heavy metals the assessment of multicomponent mixtures using TU and were focus of attention owing to their hazardous nature MTI approaches. The results of individual toxicity indi- and subsequent toxicity studies. Various workers have cated that the toxicity of Cd, Cu and Pb on P. phospho- assessed the combined toxicity of heavy metals (Su reum increased with increasing concentrations and there et al. 2012;Quetal.2013; Mochida et al. 2006). It has was a linear correlation. The 15-min IC50 values of Cd, Cu been argued that the effects caused by combinations of and Pb were 0.537, 1.905 and 1.231 mg/L, respectively, various heavy metals may be more threatening and and their toxic order was Cd [ Pb [ Cu.
    [Show full text]
  • Measuring Ligand Efficacy at the Mu- Opioid Receptor Using A
    RESEARCH ARTICLE Measuring ligand efficacy at the mu- opioid receptor using a conformational biosensor Kathryn E Livingston1,2, Jacob P Mahoney1,2, Aashish Manglik3, Roger K Sunahara4, John R Traynor1,2* 1Department of Pharmacology, University of Michigan Medical School, Ann Arbor, United States; 2Edward F Domino Research Center, University of Michigan, Ann Arbor, United States; 3Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, United States; 4Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, United States Abstract The intrinsic efficacy of orthosteric ligands acting at G-protein-coupled receptors (GPCRs) reflects their ability to stabilize active receptor states (R*) and is a major determinant of their physiological effects. Here, we present a direct way to quantify the efficacy of ligands by measuring the binding of a R*-specific biosensor to purified receptor employing interferometry. As an example, we use the mu-opioid receptor (m-OR), a prototypic class A GPCR, and its active state sensor, nanobody-39 (Nb39). We demonstrate that ligands vary in their ability to recruit Nb39 to m- OR and describe methadone, loperamide, and PZM21 as ligands that support unique R* conformation(s) of m-OR. We further show that positive allosteric modulators of m-OR promote formation of R* in addition to enhancing promotion by orthosteric agonists. Finally, we demonstrate that the technique can be utilized with heterotrimeric G protein. The method is cell- free, signal transduction-independent and is generally applicable to GPCRs. DOI: https://doi.org/10.7554/eLife.32499.001 *For correspondence: [email protected] Competing interests: The authors declare that no Introduction competing interests exist.
    [Show full text]
  • Live Biotherapeutic Products, a Road Map for Safety Assessment
    Live Biotherapeutic Products, A Road Map for Safety Assessment Alice Rouanet, Selin Bolca, Audrey Bru, Ingmar Claes, Helene Cvejic, Haymen Girgis, Ashton Harper, Sidonie Lavergne, Sophie Mathys, Marco Pane, et al. To cite this version: Alice Rouanet, Selin Bolca, Audrey Bru, Ingmar Claes, Helene Cvejic, et al.. Live Biotherapeutic Products, A Road Map for Safety Assessment. Frontiers in Medicine, Frontiers media, 2020, 7, 10.3389/fmed.2020.00237. hal-02900344 HAL Id: hal-02900344 https://hal.inrae.fr/hal-02900344 Submitted on 8 Jun 2021 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution| 4.0 International License POLICY AND PRACTICE REVIEWS published: 19 June 2020 doi: 10.3389/fmed.2020.00237 Live Biotherapeutic Products, A Road Map for Safety Assessment Alice Rouanet 1, Selin Bolca 2†, Audrey Bru 3†, Ingmar Claes 4†, Helene Cvejic 5,6†, Haymen Girgis 7†, Ashton Harper 8†, Sidonie N. Lavergne 9†, Sophie Mathys 10†, Marco Pane 11†, Bruno Pot 12,13†, Colette Shortt 14†, Wynand Alkema 15, Constance Bezulowsky 16, Stephanie Blanquet-Diot 17, Christophe Chassard 18, Sandrine P. Claus 19, Benjamin Hadida 20, Charlotte Hemmingsen 21, Cyrille Jeune 7, Björn Lindman 22, Garikai Midzi 8, Luca Mogna 11, Charlotta Movitz 22, Nail Nasir 23, 24 25 25 26 Edited by: Manfred Oberreither , Jos F.
    [Show full text]
  • Introduction
    Guidance for Assay Development & HTS March 2007 Version 5 Section I: Introduction Introduction Copyright © 2005, Eli Lilly and Company and the National Institutes of Health Chemical Genomics Center. All Rights Reserved. For more information, please review the Privacy Policy and Site Usage and Agreement. Table of Contents A. INTRODUCTION This document is written to provide guidance to investigators that are interested in developing assays useful for the evaluation of compound collections to identify chemical probes that modulate the activity of biological targets. Originally written as a guide for therapeutic projects teams within a major pharmaceutical company, this manual has been adapted to provide guidelines for: a. Identifying potential assay formats compatible with High Throughput Screen (HTS), and Structure Activity Relationship (SAR) b. Developing optimal assay reagents c. Optimizing assay protocol with respect to sensitivity, dynamic range, signal intensity and stability d. Adopting screening assays to automation and scale up in microtiter plate formats e. Statistical validation of the assay performance parameters f. Secondary follow up assays for chemical probe validation and SAR refinement g. Data standards to be followed in reporting screening and SAR assay results. General definition of biological assays This manual is intended to provide guidance in the area of biological assay development, screening and compound evaluation. In this regard an assay is defined by a set of reagents that produce a detectable signal allowing a biological process to be quantified. In general, the quality of an assay is defined by the robustness and reproducibility of this signal in the absence of any test compounds or in the presence of inactive compounds.
    [Show full text]
  • Pharmacology Part 2: Introduction to Pharmacokinetics
    J of Nuclear Medicine Technology, first published online May 3, 2018 as doi:10.2967/jnmt.117.199638 PHARMACOLOGY PART 2: INTRODUCTION TO PHARMACOKINETICS. Geoffrey M Currie Faculty of Science, Charles Sturt University, Wagga Wagga, Australia. Regis University, Boston, USA. Correspondence: Geoff Currie Faculty of Science Locked Bag 588 Charles Sturt University Wagga Wagga 2678 Australia Telephone: 02 69332822 Facsimile: 02 69332588 Email: [email protected] Foot line: Introduction to Pharmacokinetics 1 Abstract Pharmacology principles provide key understanding that underpins the clinical and research roles of nuclear medicine practitioners. This article is the second in a series of articles that aims to enhance the understanding of pharmacological principles relevant to nuclear medicine. This article will build on the introductory concepts, terminology and principles of pharmacodynamics explored in the first article in the series. Specifically, this article will focus on the basic principles associated with pharmacokinetics. Article 3 will outline pharmacology relevant to pharmaceutical interventions and adjunctive medications employed in general nuclear medicine, the fourth pharmacology relevant to pharmaceutical interventions and adjunctive medications employed in nuclear cardiology, the fifth the pharmacology related to contrast media associated with computed tomography (CT) and magnetic resonance imaging (MRI), and the final article will address drugs in the emergency trolley. 2 Introduction As previously outlined (1), pharmacology is the scientific study of the action and effects of drugs on living systems and the interaction of drugs with living systems (1-7). For general purposes, pharmacology is divided into pharmacodynamics and pharmacokinetics (Figure 1). The principle of pharmacokinetics is captured by philosophy of Paracelsus (medieval alchemist); “only the dose makes a thing not a poison” (1,8,9).
    [Show full text]
  • An Online Tool for Calculating and Mining Dose–Response Data Nicholas A
    Clark et al. BMC Cancer (2017) 17:698 DOI 10.1186/s12885-017-3689-3 SOFTWARE Open Access GRcalculator: an online tool for calculating and mining dose–response data Nicholas A. Clark1†, Marc Hafner2†, Michal Kouril3, Elizabeth H. Williams2, Jeremy L. Muhlich2, Marcin Pilarczyk1, Mario Niepel2, Peter K. Sorger2 and Mario Medvedovic1* Abstract Background: Quantifying the response of cell lines to drugs or other perturbagens is the cornerstone of pre-clinical drug development and pharmacogenomics as well as a means to study factors that contribute to sensitivity and resistance. In dividing cells, traditional metrics derived from dose–response curves such as IC50, AUC, and Emax, are confounded by the number of cell divisions taking place during the assay, which varies widely for biological and experimental reasons. Hafner et al. (Nat Meth 13:521–627, 2016) recently proposed an alternative way to quantify drug response, normalized growth rate (GR) inhibition, that is robust to such confounders. Adoption of the GR method is expected to improve the reproducibility of dose–response assays and the reliability of pharmacogenomic associations (Hafner et al. 500–502, 2017). Results: We describe here an interactive website (www.grcalculator.org) for calculation, analysis, and visualization of dose–response data using the GR approach and for comparison of GR and traditional metrics. Data can be user-supplied or derived from published datasets. The web tools are implemented in the form of three integrated Shiny applications (grcalculator, grbrowser, and grtutorial) deployed through a Shiny server. Intuitive graphical user interfaces (GUIs) allow for interactive analysis and visualization of data. The Shiny applications make use of two R packages (shinyLi and GRmetrics) specifically developed for this purpose.
    [Show full text]
  • Lack of Correlation Between in Vitro and in Vivo Studies on the Inhibitory Effects Of
    pharmaceutics Article Lack of Correlation between In Vitro and In Vivo Studies on the Inhibitory Effects of (-)-Sophoranone on CYP2C9 Is Attributable to Low Oral Absorption and Extensive Plasma Protein Binding of (-)-Sophoranone 1, 2,3, 3 3 3 Yu Fen Zheng y, Soo Hyeon Bae y , Zhouchi Huang , Soon Uk Chae , Seong Jun Jo , Hyung Joon Shim 3, Chae Bin Lee 3, Doyun Kim 3,4, Hunseung Yoo 4 and Soo Kyung Bae 3,* 1 School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 211198, China; [email protected] 2 Q-fitter, Inc., Seoul 06578, Korea; sh.bae@qfitter.com 3 College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University, Korea, Bucheon 14662, Korea; [email protected] (Z.H.); [email protected] (S.U.C.); [email protected] (S.J.J.); [email protected] (H.J.S.); [email protected] (C.B.L.); [email protected] (D.K.) 4 Life Science R&D Center, SK Chemicals, 310 Pangyo-ro, Sungnam 13494, Korea; [email protected] * Correspondence: [email protected]; Tel.: +82-2-2164-4054 These authors contributed equally to this work. y Received: 8 March 2020; Accepted: 5 April 2020; Published: 7 April 2020 Abstract: (-)-Sophoranone (SPN) is a bioactive component of Sophora tonkinensis with various pharmacological activities. This study aims to evaluate its in vitro and in vivo inhibitory potential against the nine major CYP enzymes. Of the nine tested CYPs, it exerted the strongest inhibitory effect on CYP2C9-mediated tolbutamide 4-hydroxylation with the lowest IC50 (Ki) value of 0.966 0.149 µM (0.503 0.0383 µM), in a competitive manner.
    [Show full text]
  • (2R,6R)-Hydroxynorketamine Do Not Block NMDA Receptor Function
    Antidepressant-relevant concentrations of the ketamine metabolite (2R,6R)-hydroxynorketamine do not block NMDA receptor function Eric W. Lumsdena,1, Timothy A. Troppolib,1, Scott J. Myersc, Panos Zanosd, Yasco Aracavaa, Jan Kehre,f, Jacqueline Lovettg, Sukhan Kimc, Fu-Hua Wange,f, Staffan Schmidte,f, Carleigh E. Jenned, Peixiong Yuanh, Patrick J. Morrisi, Craig J. Thomasi, Carlos A. Zarate Jr.h, Ruin Moaddelg, Stephen F. Traynelisc,2, Edna F. R. Pereiraa,j,2, Scott M. Thompsonb,d,2, Edson X. Albuquerquea,j,k,2, and Todd D. Gouldd,j,l,m,2,3 aDepartment of Epidemiology and Public Health, Division of Translational Toxicology, University of Maryland School of Medicine, Baltimore, MD 21201; bDepartment of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201; cDepartment of Pharmacology, Emory University, Atlanta, GA 30329; dDepartment of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201; eDepartment of Physiology and Pharmacology, Karolinska Institute, SE-171 77 Stockholm, Sweden; fPronexus Analytical AB, SE-167 33 Bromma, Sweden; gBiomedical Research Center, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224; hSection on the Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892; iDivision of Preclinical Innovation, National Center for Advancing Translational Sciences, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892; jDepartment of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201; kDepartment of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201; lDepartment of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201; and mVeterans Affairs Maryland Health Care System, Baltimore, MD 21201 Edited by Solomon H.
    [Show full text]
  • Investigating the Influence of Tracer Kinetics
    Molecular Pharmacology Fast Forward. Published on July 10, 2019 as DOI: 10.1124/mol.119.116764 This article has not been copyedited and formatted. The final version may differ from this version. MOL # 116764 Investigating the influence of tracer kinetics on competition-kinetic association binding assays; identifying the optimal conditions for assessing the kinetics of low affinity compounds. David A Sykes1,2, Palash Jain1,2 & Steven J Charlton1,2,3 1School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK Downloaded from 2Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK. 3 Excellerate Bioscience Ltd, Discovery Building, BioCity, Pennyfoot Street, molpharm.aspetjournals.org Nottingham, NG1 1GF, UK at ASPET Journals on October 1, 2021 1 Molecular Pharmacology Fast Forward. Published on July 10, 2019 as DOI: 10.1124/mol.119.116764 This article has not been copyedited and formatted. The final version may differ from this version. MOL # 116764 Running title: Optimizing the design of competition association assays. Addresses for correspondence: Prof Steven Charlton School of Life Sciences Queen's Medical Centre University of Nottingham Nottingham NG7 2UH Downloaded from E-mail: [email protected] molpharm.aspetjournals.org David Sykes School of Life Sciences Queen's Medical Centre University of Nottingham at ASPET Journals on October 1, 2021 Nottingham NG7 2UH E-mail: [email protected] 2 Molecular Pharmacology Fast Forward. Published on July 10, 2019 as DOI: 10.1124/mol.119.116764 This article has not been copyedited and formatted. The final version may differ from this version.
    [Show full text]