Dose-Response-Time Data Analysis: an Underexploited Trinity

Total Page:16

File Type:pdf, Size:1020Kb

Dose-Response-Time Data Analysis: an Underexploited Trinity 1521-0081/71/1/89–122$35.00 https://doi.org/10.1124/pr.118.015750 PHARMACOLOGICAL REVIEWS Pharmacol Rev 71:89–122, January 2019 Copyright © 2018 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. ASSOCIATE EDITOR: GUNNAR SCHULTE Dose-Response-Time Data Analysis: An Underexploited Trinity Johan Gabrielsson, Robert Andersson, Mats Jirstrand, and Stephan Hjorth Division of Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden (J.G.); Fraunhofer-Chalmers Centre, Gothenburg, Sweden (R.A., M.J.); Pharmacilitator AB, Vallda, Sweden (S.H.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.) Abstract. .................................................................................... 90 I. Introduction. .............................................................................. 90 II. Methods and Models ........................................................................ 92 A. What Can Be Learned from Response-Time Data Patterns? .............................. 92 B. How Is the Biophase Typically Defined?. ................................................ 93 C. The Drug “Mechanism” Function. ...................................................... 94 D. Structure of the Pharmacodynamic Model ................................................ 95 Downloaded from E. Response-Time Patterns Impact Model Structure......................................... 95 III. Case Studies . .............................................................................. 96 A. Case Study 1: Dose-Response-Time Analysis of Nociceptive Response . ................... 96 1. Background . ........................................................................ 96 2. Models, Equations, and Exploratory Analysis ......................................... 96 by guest on September 30, 2021 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. ............. 99 4. Pharmacodynamic Interpretation and Comments ..................................... 99 B. Case Study 2: Dose-Response-Time Analysis of Locomotor Stimulation. ................... 99 1. Background . ........................................................................ 99 2. Models, Equations, and Exploratory Analysis ......................................... 99 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............101 4. Pharmacodynamic Interpretation and Comments .....................................101 C. Case Study 3: Dose-Response-Time Analysis of Functional Adaptation . ...................102 1. Background . ........................................................................102 2. Models, Equations, and Exploratory Analysis .........................................102 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............103 4. Pharmacodynamic Interpretation and Comment . .....................................103 D. Case Study 4: Dose-Response-Time Analysis of Antipsychotic Effects (Brief Psychiatric Rating Scale Scores) . ..................................................................103 1. Background . ........................................................................103 2. Models, Equations, and Exploratory Analysis .........................................104 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............105 4. Pharmacodynamic Interpretation and Comments .....................................106 E. Case Study 5: Dose-Response-Time Analysis of Bacterial Count . .........................106 1. Background . ........................................................................106 2. Models, Equations, and Exploratory Analysis .........................................106 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............106 Address correspondence to: Johan Gabrielsson, Division of Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, SE-750 07 Uppsala, Sweden. E-mail: johan. [email protected] This work was funded in part through the Marie Curie Seventh Framework Programme People Initial Training Networks European Industrial Doctorate Project [Innovative Modeling for Pharmacological Advances through Collaborative Training Grant No. 316736] and by the Swedish Foundation for Strategic Research [(both to R.A. and M.J.)]. https://doi.org/10.1124/pr.118.015750. 89 90 Gabrielsson et al. 4. Pharmacodynamic Interpretation and Comments .....................................107 F. Case Study 6: Dose-Response-Time Analysis of Cortisol–Adrenocorticotropic Hormone Action...................................................................................107 1. Background . ........................................................................107 2. Models, Equations, and Exploratory Analysis .........................................107 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............107 4. Pharmacodynamic Interpretation and Comments .....................................108 G. Case Study 7: Dose-Response-Time Analysis of Miotic Effects.............................108 1. Background . ........................................................................108 2. Models, Equations, and Exploratory Analysis .........................................109 3. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............109 4. Pharmacodynamic Interpretation and Comments .....................................109 H. Case Study 8: Meta-Analysis of Dose-Response-Time Data. ...............................110 1. Background . ........................................................................110 2. Results and Conclusions with Respect to Dose-Response-Time Analysis. .............110 3. Pharmacodynamic Interpretation and Comments .....................................111 I. Overall Conclusions of the Case Studies. ................................................111 IV. Discussion ..................................................................................112 A. Background .............................................................................112 B. What Do Different Doses, Routes, and Rates of Input Add? ...............................112 C. Drug Delivery by Means of Iontophoresis ................................................119 D. Permutations of Smolen’s Model: The Kinetic-dynamic K-PD Rate of Change Model.......120 E. General Conclusions . ..................................................................120 References ..................................................................................120 Abstract——Themostcommonapproachtoinvivo direct assay method for the drug and has the pharmacokinetic and pharmacodynamic analyses additional advantage of being applicable to cases involves sequential analysis of the plasma concen- of local drug administration close to its intended tration- and response-time data, such that the targets in the immediate vicinity of target, or when plasma kinetic model provides an independent func- target precedes systemic plasma concentrations. tion, driving the dynamics. However, in situations This review exemplifies the potential of biophase when plasma sampling may jeopardize the effect functions in pharmacodynamic analyses in both measurements or is scarce, nonexistent, or unlinked preclinical and clinical studies, with the purpose to the effect (e.g., in intensive care units, pediatric of characterizing response data and optimizing or frail elderly populations, or drug discovery), subsequent study protocols. This article illustrates focusing on the response-time course alone may be crucial determinants to the success of modeling an adequate alternative for pharmacodynamic analyses. dose-response-time (DRT) data, such as the dose Response-time data inherently contain useful information selection, repeated dosing, and different input about the turnover characteristics of response (target rates and routes. Finally, a literature search was also turnover rate, half-life of response), as well as the drug’s performed to gauge how frequently this technique has biophase kinetics (biophase availability, absorption been applied in preclinical and clinical studies. This half-life, and disposition half-life) pharmacodynamic review highlights situations in which DRT should be properties (potency, efficacy). The use of pharmacodynamic carefully scrutinized and discusses future perspectives time-response data circumvents the need for a of the field. I. Introduction pharmacological response when information about the drug concentration is scarce or even totally lacking Pharmacological time-series data are typically rich in (Levy, 1971; Smolen, 1971a,b, 1976a,b, 1978; Smolen information. However, response-time series are often et al., 1972; Smolen and Weigand, 1973). Usually, compared with plasma or blood exposure prior to any plasma or tissue concentrations of drug are used as analysis of the connection between dose, response, and the “driver” of the pharmacological response. However, time data per se. Dose-response-time (DRT) data anal- plasma concentrations may be of marginal value ysis is therefore an underexplored approach for quan- in situations with difficult or unethical sampling (e.g., tification of the onset, intensity,
Recommended publications
  • 1: Clinical Pharmacokinetics 1
    1: CLINICAL PHARMACOKINETICS 1 General overview: clinical pharmacokinetics, 2 Pharmacokinetics, 4 Drug clearance (CL), 6 Volume of distribution (Vd), 8 The half-life (t½), 10 Oral availability (F), 12 Protein binding (PB), 14 pH and pharmacokinetics, 16 1 Clinical pharmacokinetics General overview General overview: clinical pharmacokinetics 1 The ultimate aim of drug therapy is to achieve effi cacy without toxicity. This involves achieving a plasma concentration (Cp) within the ‘therapeutic window’, i.e. above the min- imal effective concentration (MEC), but below the minimal toxic concentration (MTC). Clinical pharmacokinetics is about all the factors that determine variability in the Cp and its time-course. The various factors are dealt with in subsequent chapters. Ideal therapeutics: effi cacy without toxicity Minimum Toxic Concentration (MTC) Ideal dosing Minimum Effective Concentration (MEC) Drug concentration Time The graph shows a continuous IV infusion at steady state, where the dose-rate is exactly appropriate for the patient’s clearance (CL). Inappropriate dosing Dosing too high in relation to the patient’s CL – toxicity likely Minimum Toxic Concentration (MTC) Minimum Effective Concentration (MEC) Dosing too low in relation to the Drug concentration patient’s CL – drug may be ineffective Time Some reasons for variation in CL Low CL High CL Normal variation Normal variation Renal impairment Increased renal blood fl ow Genetic poor metabolism Genetic hypermetabolism Liver impairment Enzyme induction Enzyme inhibition Old age/neonate 2 General overview Clinical Pharmacokinetics Pharmacokinetic factors determining ideal therapeutics If immediate effect is needed, a loading dose (LD) must be given to achieve a desired 1 concentration. The LD is determined by the volume of distribution (Vd).
    [Show full text]
  • Prediction of Clinical Transporter‐Mediated Drug–Drug Interactions
    Citation: CPT Pharmacometrics Syst. Pharmacol. (2020) 9, 211–221; doi:10.1002/psp4.12505 ARTICLE Prediction of Clinical Transporter-Mediated Drug–Drug Interactions via Comeasurement of Pitavastatin and Eltrombopag in Human Hepatocyte Models Simon J. Carter1, Bhavik Chouhan2, Pradeep Sharma3 and Michael J. Chappell1,* A structurally identifiable micro-rate constant mechanistic model was used to describe the interaction between pitavastatin and eltrombopag, with improved goodness-of-fit values through comeasurement of pitavastatin and eltrombopag. Transporter association and dissociation rate constants and passive rates out of the cell were similar between pitavastatin and eltrom- bopag. Translocation into the cell through transporter-mediated uptake was six times greater for pitavastatin, leading to pronounced inhibition of pitavastatin uptake by eltrombopag. The passive rate into the cell was 91 times smaller for pitavas- tatin compared with eltrombopag. A semimechanistic physiologically-based pharmacokinetic (PBPK) model was developed to evaluate the potential for clinical drug–drug interactions (DDIs). The PBPK model predicted a twofold increase in the pita- vastatin peak blood concentration and area under the concentration-time curve in the presence of eltrombopag in simulated healthy volunteers. The use of structural identifiability supporting experimental design combined with robust micro-rate con- stant parameter estimates and a semimechanistic PBPK model gave more informed predictions of transporter-mediated DDIs. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? model fits through micro-rate constants compared with ✔ Currently, most in vitro models are not guided by macro-rate constants in human hepatocytes, with addi- structural identifiability analysis, relying on substrate-only tional information provided regarding transporter binding.
    [Show full text]
  • Using Toxicity Tests in Ecological Risk Assessment Toxicity Tests Are Used to Expose Test Organisms to a 2
    United States Office of Publication 9345.0-05I Environmental Solid Waste and March 1994 Protection Emergency Response Agency ECO Update Office of Emergency Remedial Response Intermittent Bulletin Hazardous Site Evaluation Division (5204G) Volume 2 Number1 Using Toxicity Tests in Ecological Risk Assessment Toxicity tests are used to expose test organisms to a 2. Toxicity tests can evaluate the aggregate toxic effects of medium—water, sediment, or soil—and evaluate the effects of all contaminants in a medium. Many Superfund sites present contamination on the survival, growth, reproduction, behavior a complex array of contaminants, with a mixture of potentially and/or other attributes of these organisms. These tests may harmful substances present in the media. At such sites, help to determine whether the contaminant concentrations in a chemical data alone cannot accurately predict the toxicity of site’s media are high enough to cause adverse effects in the contaminants. Rather, toxicity tests measure the aggregate organisms. Generally, toxicity tests involve collecting effects of contaminated media on organisms. These effects samples of media from a site and sending them to a toxicity result from characteristics of the medium itself (such as laboratory, where the tests are performed. On occasion hardness and pH, in the case of water), interactions among investigators1 measure toxicity by exposing test organisms to contaminants, and interactions between contaminants and soil or water on site—these are known as in situ tests. media. Consequently, observed toxicity test results may often vary from those predicted by chemical data alone. As the general guidelines at the end of this Bulletin indicate, not all sites require toxicity tests.
    [Show full text]
  • Gene-Signature-Derived Ic50s/Ec50s Reflect the Potency of Causative
    www.nature.com/scientificreports OPEN Gene-signature-derived IC50s/EC50s refect the potency of causative upstream targets and downstream phenotypes Stefen Renner1 ✉ , Christian Bergsdorf1, Rochdi Bouhelal1, Magdalena Koziczak-Holbro2, Andrea Marco Amati1,6, Valerie Techer-Etienne1, Ludivine Flotte2, Nicole Reymann1, Karen Kapur3, Sebastian Hoersch3, Edward James Oakeley4, Ansgar Schufenhauer1, Hanspeter Gubler3, Eugen Lounkine5,7 & Pierre Farmer1 ✉ Multiplexed gene-signature-based phenotypic assays are increasingly used for the identifcation and profling of small molecule-tool compounds and drugs. Here we introduce a method (provided as R-package) for the quantifcation of the dose-response potency of a gene-signature as EC50 and IC50 values. Two signaling pathways were used as models to validate our methods: beta-adrenergic agonistic activity on cAMP generation (dedicated dataset generated for this study) and EGFR inhibitory efect on cancer cell viability. In both cases, potencies derived from multi-gene expression data were highly correlated with orthogonal potencies derived from cAMP and cell growth readouts, and superior to potencies derived from single individual genes. Based on our results we propose gene-signature potencies as a novel valid alternative for the quantitative prioritization, optimization and development of novel drugs. Gene expression signatures are widely used in the feld of translational medicine to defne disease sub-types1, severity2 and predict treatment outcome3. Bridging this technology to early drug discovery was previously pro- posed years ago4,5 but its prohibitive costs limited this approach. Te recent advancement of massively parallel gene expression technologies such as RASL-seq.6, DRUG-seq.7, QIAseq.8,9, PLATE-seq.10, or LINCS L100011 are now transforming the feld of compound profling, enabling larger scale profling and screening experiments at a more afordable cost12–17.
    [Show full text]
  • Development of Macromolecular Prodrugs for the Treatment of Chronic Inflammatory Pain
    University of Nebraska Medical Center DigitalCommons@UNMC Theses & Dissertations Graduate Studies Fall 12-14-2018 Development of Macromolecular Prodrugs for the Treatment of Chronic Inflammatory Pain Laura Weber University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Pharmacy and Pharmaceutical Sciences Commons Recommended Citation Weber, Laura, "Development of Macromolecular Prodrugs for the Treatment of Chronic Inflammatory Pain" (2018). Theses & Dissertations. 323. https://digitalcommons.unmc.edu/etd/323 This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected]. DEVELOPMENT OF MACROMOLECULAR PRODRUGS FOR THE TREATMENT OF CHRONIC INFLAMMATORY PAIN By Laura Weber A DISSERTATION Presented to the Faculty of The Graduate College of the University of NeBraska In Partial Fulfillment of the Requirements For the Degree of Doctor of Philosophy Department of Pharmaceutical Sciences Under the Supervision of Professor Dong Wang University of NeBraska Medical Center Omaha, NeBraska DecemBer 2018 TABLE OF CONTENTS ACKNOWLEDGEMENTS .............................................................................................. I ABSTRACT ................................................................................................................. IV LIST OF
    [Show full text]
  • 2D6 Substrates 2D6 Inhibitors 2D6 Inducers
    Physician Guidelines: Drugs Metabolized by Cytochrome P450’s 1 2D6 Substrates Acetaminophen Captopril Dextroamphetamine Fluphenazine Methoxyphenamine Paroxetine Tacrine Ajmaline Carteolol Dextromethorphan Fluvoxamine Metoclopramide Perhexiline Tamoxifen Alprenolol Carvedilol Diazinon Galantamine Metoprolol Perphenazine Tamsulosin Amiflamine Cevimeline Dihydrocodeine Guanoxan Mexiletine Phenacetin Thioridazine Amitriptyline Chloropromazine Diltiazem Haloperidol Mianserin Phenformin Timolol Amphetamine Chlorpheniramine Diprafenone Hydrocodone Minaprine Procainamide Tolterodine Amprenavir Chlorpyrifos Dolasetron Ibogaine Mirtazapine Promethazine Tradodone Aprindine Cinnarizine Donepezil Iloperidone Nefazodone Propafenone Tramadol Aripiprazole Citalopram Doxepin Imipramine Nifedipine Propranolol Trimipramine Atomoxetine Clomipramine Encainide Indoramin Nisoldipine Quanoxan Tropisetron Benztropine Clozapine Ethylmorphine Lidocaine Norcodeine Quetiapine Venlafaxine Bisoprolol Codeine Ezlopitant Loratidine Nortriptyline Ranitidine Verapamil Brofaramine Debrisoquine Flecainide Maprotline olanzapine Remoxipride Zotepine Bufuralol Delavirdine Flunarizine Mequitazine Ondansetron Risperidone Zuclopenthixol Bunitrolol Desipramine Fluoxetine Methadone Oxycodone Sertraline Butylamphetamine Dexfenfluramine Fluperlapine Methamphetamine Parathion Sparteine 2D6 Inhibitors Ajmaline Chlorpromazine Diphenhydramine Indinavir Mibefradil Pimozide Terfenadine Amiodarone Cimetidine Doxorubicin Lasoprazole Moclobemide Quinidine Thioridazine Amitriptyline Cisapride
    [Show full text]
  • 624.Full.Pdf
    1521-009X/44/5/624–633$25.00 http://dx.doi.org/10.1124/dmd.115.068932 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 44:624–633, May 2016 Copyright ª 2016 by The American Society for Pharmacology and Experimental Therapeutics Development of a Rat Plasma and Brain Extracellular Fluid Pharmacokinetic Model for Bupropion and Hydroxybupropion Based on Microdialysis Sampling, and Application to Predict Human Brain Concentrations Thomas I.F.H. Cremers, Gunnar Flik, Joost H.A. Folgering, Hans Rollema, and Robert E. Stratford, Jr. Brains On-Line BV, Groningen, The Netherlands (T.I.F.H.C., G.F. J.H.A.F.); Rollema Biomedical Consulting, Mystic, Connecticut (H.R.); and Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (R.E.S.) Received December 11, 2015; accepted February 24, 2016 Downloaded from ABSTRACT Administration of bupropion [(6)-2-(tert-butylamino)-1-(3-chlorophenyl) concentration ratio at early time points was observed with propan-1-one] and its preformed active metabolite, hydroxybupropion bupropion; this was modeled as a time-dependent uptake clear- [(6)-1-(3-chlorophenyl)-2-[(1-hydroxy-2-methyl-2-propanyl)amino]- ance of the drug across the blood–brain barrier. Translation of the 1-propanone], to rats with measurement of unbound concentrations model was used to predict bupropion and hydroxybupropion dmd.aspetjournals.org by quantitative microdialysis sampling of plasma and brain extra- exposure in human brain extracellular fluid after twice-daily cellular fluid was used to develop a compartmental pharmacoki- administration of 150 mg bupropion. Predicted concentrations netics model to describe the blood–brain barrier transport of both indicate that preferential inhibition of the dopamine and norepi- substances.
    [Show full text]
  • Clinical Pharmacokinetics 38
    Clin Pharmacokinet 2000 Jun; 38 (6): 505-518 ORIGINAL RESEARCH ARTICLE 0312-5963/00/0006-0505/$20.00/0 © Adis International Limited. All rights reserved. Pharmacokinetic-Pharmacodynamic Modelling of the Antipyretic Effect of Two Oral Formulations of Ibuprofen Iñaki F. Trocóniz,1 Santos Armenteros,2 María V. Planelles,3 Julio Benítez,4 Rosario Calvo5 and Rosa Domínguez2 1 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Navarra, Pamplona, Spain 2 Medical Department, Laboratorios Knoll S.A., Madrid, Spain 3 Department of Paediatrics, Clinic Hospital, Valencia, Spain 4 Department of Pharmacology, Faculty of Medicine, University of Extremadura, Badajoz, Spain 5 Department of Pharmacology, Faculty of Medicine, University of the Basque Country, Lejona, Spain Abstract Objective: To analyse the population pharmacokinetic-pharmacodynamic relation- ships of racemic ibuprofen administered in suspension or as effervescent granules with the aim of exploring the effect of formulation on the relevant pharmaco- dynamic parameters. Design: The pharmacokinetic model was developed from a randomised, cross- over bioequivalence study of the 2 formulations in healthy adults. The pharmaco- dynamic model was developed from a randomised, multicentre, single dose efficacy and safety study of the 2 formulations in febrile children. Patients and participants: Pharmacokinetics were studied in 18 healthy volun- teers aged 18 to 45 years, and pharmacodynamics were studied in 103 febrile children aged between 4 and 16 years with bodyweight ≥25kg. Methods: The pharmacokinetic study consisted of two 1-day study occasions, each separated by a 1-week washout period. On each occasion ibuprofen 400mg was administered orally as suspension or granules. The time course of the anti- pyretic effect was evaluated in febrile children receiving a single oral dose of 7 mg/kg in suspension or 200 or 400mg as effervescent granules.
    [Show full text]
  • Antimicrobial Stewardship Guidance
    Antimicrobial Stewardship Guidance Federal Bureau of Prisons Clinical Practice Guidelines March 2013 Clinical guidelines are made available to the public for informational purposes only. The Federal Bureau of Prisons (BOP) does not warrant these guidelines for any other purpose, and assumes no responsibility for any injury or damage resulting from the reliance thereof. Proper medical practice necessitates that all cases are evaluated on an individual basis and that treatment decisions are patient-specific. Consult the BOP Clinical Practice Guidelines Web page to determine the date of the most recent update to this document: http://www.bop.gov/news/medresources.jsp Federal Bureau of Prisons Antimicrobial Stewardship Guidance Clinical Practice Guidelines March 2013 Table of Contents 1. Purpose ............................................................................................................................................. 3 2. Introduction ...................................................................................................................................... 3 3. Antimicrobial Stewardship in the BOP............................................................................................ 4 4. General Guidance for Diagnosis and Identifying Infection ............................................................. 5 Diagnosis of Specific Infections ........................................................................................................ 6 Upper Respiratory Infections (not otherwise specified) ..............................................................................
    [Show full text]
  • CPIC Guideline for Pharmacogenetics-Guided Warfarin Dosing – Supplement V2.0 1 Table of Contents Guideline Updates
    Supplement to: Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for Pharmacogenetics-guided Warfarin Dosing: 2016 Update Julie A. Johnson1, Kelly Caudle2, Li Gong3, Michelle Whirl-Carrillo3, C. Michael Stein4, Stuart A. Scott5, Ming Ta Michael Lee6 , Brian F. Gage7, Stephen E. Kimmel8,9, Minoli A. Perera10, Jeffrey L. Anderson11, Munir Pirmohamed12, Teri E. Klein3, Nita A. Limdi13, Larisa H. Cavallari1, Mia Wadelius14 1Department of Pharmacotherapy and Translational Research, College of Pharmacy, and Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA 2Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 3Department of Genetics, Stanford University, Stanford, California, USA 4Division of Clinical Pharmacology Vanderbilt Medical School, Nashville, TN, USA 5Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA 6Laboratory for International Alliance on Genomic Research, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; National Center for Genome Medicine; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Genomic Medicine Institute Geisinger Health system, Danville, PA 7Department of Internal Medicine, Washington University in St. Louis, St. Louis, Missouri 8Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA 9Department of Medicine and Department of Biostatistics and Epidemiology, University of Pennsylvania
    [Show full text]
  • Emergency Use Authorization (EUA) for Bamlanivimab 700 Mg and Etesevimab 1400 Mg IV Administered Together Center for Drug Evaluation and Research (CDER) Review
    ________________________________________________________________ Emergency Use Authorization (EUA) for Bamlanivimab 700 mg and Etesevimab 1400 mg IV Administered Together Center for Drug Evaluation and Research (CDER) Review Identifying Information Application Type (EUA or Pre-EUA) EUA If EUA, designate whether pre-event or intra-event EUA request. 1 EUA Application Number(s) EUA 000094 Sponsor (entity requesting EUA or Eli Lilly and Company pre-EUA consideration), point of Christine Phillips, PhD, RAC contact, address, phone number, fax Advisor, Global Regulatory Affairs - NA (b) (6) number, email address Mobile: Email: [email protected] Manufacturer, if different from Eli Lilly and Company Sponsor Submission Date(s) November 16, 2020 Receipt Date(s) November 16, 2020 OND Division / Office Division of Antivirals/Office of Infectious Diseases Reviewer Name(s)/Discipline(s) Natalie Pica, MD, PhD – Clinical Reviewer Wendy Carter, DO – Cross Discipline Team Lead Michael Thomson, PhD – Clinical Virology Reviewer Jules O’Rear, PhD – Clinical Virology Team Lead Daniel Rubin, PhD – Statistics Reviewer Thamban Valappil, PhD – Statistics Team Lead Dionne Price, PhD – Director, Division of Biometrics IV Jason Moore, PharmD – Clinical Pharmacology Reviewer Su-Young Choi PharmD, PhD – Clinical Pharmacology Team Lead Kellie Reynolds, PharmD – Division Director, DIDP Ye Xiong, PhD – Pharmacometrics Reviewer Justin Earp, PhD – Pharmacometrics Team Lead Yaning Wang, PhD – Division Director, DPM 1 If a Pre-EUA is in existence at the time
    [Show full text]
  • Efavirenz) Capsules and Tablets 3 Rx Only
    1 SUSTIVA® 2 (efavirenz) capsules and tablets 3 Rx only 4 DESCRIPTION 5 SUSTIVA® (efavirenz) is a human immunodeficiency virus type 1 (HIV-1) specific, non- 6 nucleoside, reverse transcriptase inhibitor (NNRTI). 7 Capsules: SUSTIVA is available as capsules for oral administration containing either 8 50 mg, 100 mg, or 200 mg of efavirenz and the following inactive ingredients: lactose 9 monohydrate, magnesium stearate, sodium lauryl sulfate, and sodium starch glycolate. 10 The capsule shell contains the following inactive ingredients and dyes: gelatin, sodium 11 lauryl sulfate, titanium dioxide, and/or yellow iron oxide. The capsule shells may also 12 contain silicon dioxide. The capsules are printed with ink containing carmine 40 blue, 13 FD&C Blue No. 2, and titanium dioxide. 14 Tablets: SUSTIVA is available as film-coated tablets for oral administration containing 15 600 mg of efavirenz and the following inactive ingredients: croscarmellose sodium, 16 hydroxypropyl cellulose, lactose monohydrate, magnesium stearate, microcrystalline 17 cellulose, and sodium lauryl sulfate. The film coating contains Opadry® Yellow and 18 Opadry® Clear. The tablets are polished with carnauba wax and printed with purple ink, 19 Opacode® WB. 20 Efavirenz is chemically described as (S)-6-chloro-4-(cyclopropylethynyl)-1,4-dihydro-4- 21 (trifluoromethyl)-2H-3,1-benzoxazin-2-one. 22 Its empirical formula is C14H9ClF3NO2 and its structural formula is: 1 of 45 Approved v2.0 F C 3 Cl O NO 23 H 24 Efavirenz is a white to slightly pink crystalline powder with a molecular mass of 315.68. 25 It is practically insoluble in water (<10 µg/mL).
    [Show full text]