PREDICTING PHARMACOKINETICS FOLLOWING TOPICAL APPLICATION USING NON-ANIMAL METHODS IAN SORRELL, MI-YOUNG LEE, RICHARD CUBBERLEY

Similar documents
XTreme 200 Human Liver Microsomes Lot No Human Liver Microsomes Pool of 200 (100 Male and 100 Female) Suspension medium: 250 mm sucrose

Chris Bohl, Ph.D. Global Technical Support Manager- Products

Minireview. Low-Turnover Drug Molecules: A Current Challenge for Drug Metabolism Scientists

Culture Hepatocytes in Human Plasma to Count the free Concentration of Drug in Evaluation of Drug-drug Interaction. Chuang Lu

Development & Characterization of Pooled and Plated Hepatocytes to Support the Evolving DMPK Landscape

Effect of BD Matrigel Matrix Overlay and BD Matrigel Matrix Thin Coat on CYP450 Activities in Cryo Human Hepatocytes. Rongjun Zuo.

CASE STUDY PRESENTATION: A QUANTITATIVE AOP FOR SKIN SENSITISATION RISK ASSESSMENT

Metabolism. Objectives. Metabolism. 26 July Chapter 28 1

Bioprinted 3D Primary Liver Tissues. PD Dr Adrian Roth Head Mechanistic Safety Roche Innovation Centre Basel, Switzerland

RSC, February Interplay between enzymes and. clearance and intracellular concentration of drugs. Centre for Applied Pharmacokinetic Research

FROM PATHWAYS TO PEOPLE: APPLYING THE SKIN SENSITISATION AOP TO RISK ASSESSMENT

Comparison Between the US FDA, Japan PMDA and EMA In Vitro DDI Guidance: Are we Close to Harmonization?

Cryo Characterization Report (CCR)

In Vitro In Vivo Extrapolation (IVIVE): Why It Is Not As Easy As You May Think

Evaluation of Cryopreserved Human Hepatocytes as an Alternative In Vitro System to

Involvement of CYP2C8 and UGT1A9 in the metabolism of a novel gastroprokinetic agent, Z-338

It the process by which a drug reversibly leaves blood and enter interstitium (extracellular fluid) and/ or cells of tissues.

The Future of In Vitro Systems for the Assessment of Induction and Suppression of Enzymes and Transporters

Assessing the role of hepatic uptake in drug clearance - Pharmacokinetic and experimental considerations

DMPK. APRIL 27 TH 2017 Jan Neelissen Scientific Adviser Science & Technology

Michelle Schaefer, Gerhard Schänzle, Daniel Bischoff, and Roderich D. Süssmuth.

Skin Sensitization MoA/AOP pathway elucidation: Applying the Skin Sensitization AOP to Risk Assessment

Under prediction of hepatic clearance from in vitro studies: prospects for resolution. J Brian Houston

Prediction of DDIs Arising from CYP3A Induction Using a Physiologically-based Dynamic Model. Lisa Almond 22 nd June 2016

Basic Concepts in Pharmacokinetics. Leon Aarons Manchester Pharmacy School University of Manchester

3/20/2007 Page Mechanisms of Drug Action. The Liver and Metabolism September 30, 2005

Strategies for In Vitro Metabolic Stability Testing. Christopher Patten, PhD. BD Biosciences December 2, 2009

Stimulate your kinetic understanding Permeability Binding Metabolism Transporters

Carolina Säll, J. Brian Houston, and Aleksandra Galetin

In Vitro ADMET Laboratories Inc. A Hepatocyte Enterocyte Research Organization. Columbia, MD and Malden, MA

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

University of Groningen. Human and rat organ slices de Kanter, Ruben

DILI: Clinical Pharmacology Considerations for Risk Assessment

Muhammad Fawad Rasool Feras Khalil Stephanie Läer

Chapter 4. Drug Biotransformation

Click to edit Master title style

NEXT GENERATION RISK ASSESSMENT FOR CONSUMER SAFETY OF COSMETICS: A CASE STUDY APPROACH

Section 5.2: Pharmacokinetic properties

Current Approaches and Applications of Phenotyping Methods for Drug Metabolizing Enzymes and Transporters

NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES S3A

An In Vitro Alternative For Predicting Systemic Toxicity. By: James McKim, Ph.D., DABT Chief Science Officer

DEVELOPMENTAL PK/PD: WHAT HAVE WE LEARNT? Geoff Tucker

Optimisation of In Vitro Methodology for Drug Metabolism Studies to Improve Prediction of Hepatic Drug Clearance

Pharmacokinetic Modeling & Simulation in Discovery and non-clinical Development

Putting Science to Work. Heptox Virtual Liver Platform

III. TOXICOKINETICS. Studies relevant to the toxicokinetics of inorganic chloramines are severely

Toxicant Disposition and Metabolism. Jan Chambers Center for Environmental Health Sciences College of Veterinary Medicine

Introduction to Pharmacokinetics (PK) Anson K. Abraham, Ph.D. Associate Principal Scientist, PPDM- QP2 Merck & Co. Inc., West Point, PA 5- June- 2017

Drug disposition classification systems: A comparative review of BDDCS, ECCS and ECCCS

Novel Hepatocyte Technologies for the Evaluation of Adverse Drug Properties

Cryopreserved Enterocytes for the Evaluation of Drug-Drug and Food- Drug Interactions

Industrial Toxicology

Investigations of Bi-phasic Drug Depletion in Liver Microsomes and Hepatocytes in Metabolic Stability Studies

Mechanism of Action of N-Acetylcysteine in the Protection Against the Hepatotoxicity of Acetaminophen in Rats In Vivo

Influence of fluvoxamine on carvedilol metabolism and plasma disposition in vitro and in vivo experiments

Cytochrome P450 Suppression in Human Hepatocyte Cultures by Small and Large Molecules. George Zhang, Ph.D. April 18, 2012

Automation of TRANSIL assays outperforms. terms of speed, cost-effectiveness and reproducibility. Dr. Hinnerk Boriss

Cryopreserved HepaRG Cells and Media Supplements

Renal Function. 1. Glomerular filtration 2. Active tubular secretion 3. Passive tubular reabsorption 4. Excretion

Importance of Multi-P450 Inhibition in Drug Drug Interactions: Evaluation of Incidence, Inhibition Magnitude, and Prediction from in Vitro Data

Drug Interactions, from bench to bedside

Metabolic Changes of Drugs and Related Organic Compounds

Drug Metabolism Phase 2 conjugation reactions. Medicinal chemistry 3 rd stage

HTPK: Conducting PK modeling and

Caveat: Validation and Limitations of Phenotyping Methods for Drug Metabolizing Enzymes and Transporters

Elizabeth Crawford 1, Shaoxia Yu 2, Lawrence Cohen 2 Justin Gordon 2, Brian Musselman 1, Jing-Tao Wu 2. IonSense, Inc., Saugus, MA

Pharmacokinetics of Drugs. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

Strategy on Drug Transporter Investigation Why, How, Which & When. Jasminder Sahi

Effects of Liver Disease on Pharmacokinetics Juan J.L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program October 29, 2015 National

Effects of Liver Disease on Pharmacokinetics Juan J.L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program November 4, 2010 National

PBPK modeling of renal impairment what is missing?

Opportunities and Challenges for Using IVIVE to Improve Decision Making. Weihsueh A. Chiu, PhD Texas A&M University

The liver in poisoning: what can we learn from animal models?

FDA s Clinical Drug Interaction Studies Guidance (2017 Draft Guidance)

The metabolism of 1 - and 4-hydroxymidazolam by glucuronide conjugation is. largely mediated by UDP-glucuronosyltransferases 1A4, 2B4, and 2B7

Characterization of in vitro glucuronidation clearance of a range of drugs in human kidney

BRIDGE THE GAP A Human Pathways Approach to Disease Research

General Pharmacology MCQs

Physiologically Based Pharmacokinetic Modeling to Predict Drug-Drug Interactions. Involving Inhibitory Metabolite A Case Study of Amiodarone

The Importance of ADME/PK to Inform Human Safety Assessments Based on Animal Studies: Example with Furan. Gregory L. Kedderis, PhD Chapel Hill, NC

COPYRIGHTED MATERIAL IN VITRO EVALUATION OF METABOLIC DRUG DRUG INTERACTIONS: CONCEPTS AND PRACTICE ALBERT P. LI

Basic pharmacokinetics. Frédérique Servin APHP hôpital Bichat Paris, FRANCE

Novel Human-Based In Vitro Liver and Intestinal Technologies for Drug Development

Can PK and Modelling Help?

Hepatotoxicity Test by Stem Cell derived Hepatocyte

Presentation of the SEURAT-1 COSMOS Project: Prediction of Systemic Toxicity Following Dermal Exposure

HISTOPL4THOLOG1CAL STUDY OF LIVER

DRUG ELIMINATION II BILIARY EXCRETION MAMMARY, SALIVARY AND PULMONARY EXCRETION

B. Incorrect! Compounds are made more polar, to increase their excretion.

Hepatocyte Metabolic Kinetics Assays

Helping the liver to detoxify mycotoxins

Guidelines/Supporting Studies* FDA Label Information Additional Information/Commentsxc` Gene(s)/Level of evidence

In vitro metabolism of montelukast by Cytochrome P450s (CYPs) and UDPglucuronosyltransferases

Brainteaser NK-1 receptor antagonists

3D Tissue Models. Simple, Low Cost Fabrication. Simple, Robust Protocols

Introduction to Pharmacokinetics

Evaluation of Drug-Drug Interactions FDA Perspective

Enhancement of Physiological Relevance of In Vitro Assays

Chuang Lu, Suresh K. Balani, Mark G. Qian, Shimoga R. Prakash, Patricia S. Ducray, and Lisa L. von Moltke

Incorporating Gene-Environment Information into Kinetic Models: Lessons Learned and Future Challenges

Transcription:

PREDICTING PHARMACOKINETICS FOLLOWING TOPICAL APPLICATION USING NON-ANIMAL METHODS IAN SORRELL, MI-YOUNG LEE, RICHARD CUBBERLEY

UNILEVER APPLICATIONS KINETICS Need for estimates of local and systemic concentrations of chemical ingredients Efficacy Toxicology Use of kinetics in risk assessments without the use of animal models: Predict internal concentrations with quantified uncertainty IVIVE Support first in man Safety decision making

PREDICTING INTERNAL CONCENTRATIONS Systemic Exposure In Vitro Assays: Kinetic Solubility Thermodynamic Solubility Metabolic Stability -Human Hepatocytes -Human CYP450 Isoforms -Human Hepatic Microsomes Stability in Human Plasma Plasma Protein Binding Partitioning in Human Blood Gastroplus SimulationsPlus Predicting systemic exposure Enabling us to select and test relevant doses Increased role for clinical work to confirm systemic exposure levels PBK Modelling

EVALUATION OF PBK PREDICTIONS FOR TOPICAL APPLICATION ESTIMATING UNCERTAINTY IN PREDICTIONS OF SYSTEMIC CONCENTRATION

PREDICTION VS. OBSERVATION Comparing predicted Cmax with measured clinical data (plasma concentration time profile following topical application) Limited by number compounds Skin penetration in vitro Metabolism hepatocyte suspension Experimental measurements protein binding Gastroplus PBK model

Skin Bioavailabity DERMAL PENETRATION Dermal Kinetics ex vivo human skin Dealing with uncertainty: Bayesian inference Understanding the kinetics of an ingredient in the skin to allow risk assessments for local endpoints Understanding delivery to the systemic circulation following dermal application Davies et al (2011) Toxicol Sci 119, 308-18

EVALUATION IN VITRO HEPATIC METABOLISM ASSAYS METHODS FOR LOW CLEARANCE COMPOUNDS

HEPATIC CLEARANCE Current models for human metabolism Subcellular fraction Easy to use Express phase I/II enzyme Used for clearance, inhibition, covalent binding Pool of large number of donors Can be recovered from frozen tissues Cell line: Lower cost, renewable source Need to be evaluated for target application Cell culture / cell line Easy to use: amenable to high throughput platform Used for toxicity & DDI studies Co-culture with other cells Able to be cryopreserved Available for multiple species De-differentiate in culture Lack representation of intact liver structure Cell line: Lower cost, renewable source Need to be evaluated for target application 3D cell culture Allow better cell-cell & cell-matrix interactions Represent the in vivo-like conditions in terms of cell function, morphology, nutrition, oxygenation, configuration Enabling better prediction of drug toxicity Maintain long term culture; good for chronic drug treatment, slow clearance drug metabolism Low to medium throughput Some 3D formats difficult for imaging Liver cut slices In vivo architecture reserved In vivo-like expression of drug metabolizing enzymes, transporters, and functional bile canaliculi Zone specific metabolism in toxicity may be studied Hepatic function reserved for <24 hrs. Complicated to use Difficult to obtain human tissue Complexity, Ability to predict human metabolism, In Vivo systems resembling, Cost Easy of handling, Reproducibility In Vivo models Animal models not used by Unilever Have inter-species differences High cost Most suitable model to study different organinteraction In Vitro-In Vivo extrapolation Use of in vitro models to predict in vivo information

IN VITRO STUDY OF LOW CLEARANCE CHEMICALS IN VITRO SYSTEMS FOR METABOLISM STUDY Non-cell Cell, primary human hepatocytes preparation Human liver fraction Mono culture Co-culture Types S9 microsomes Suspended culture Plated culture Hanging-drop microtissue Micropatterned culture Incubation time (hr) 2 2 4 24 24-72 (168) 168 Increase incubation time Read-out Drug depletion rates Metabolite profile Cell viability In single concentration (1µM), In vitro clearance (to be extrapolated to in vivo CL) Metabolite formation and possible metabolic pathway (Quantitative and qualitative data) Cell viability spheroid assay during 7 days incubation

SPHEROID AND MICROPATTERNED HEPATOCYTES Spheroids from InSphero Co-culture: Pooled Primary Human Hepatocyte (PHHs) 10 mixed donors Kupffer cells -1 donor Cell number: total 1,000 cells per spheroid, 500 hepatocytes/ spheroid Microtissue size: 225.7 ± 11.1 µm Incubation vol.: 96 well (100 microliters / well) Function: Stable liver function until 4 weeks Solvo Biotechnology Ascendance: HepatoPac External work Co-culture: Primary Human Hepatocyte (PHHs) single donor 3121B 3T3 stromal cells mouse cell line Cell number: 3,200 cells (Hepatocytes), 15,000 cells (stromal) Incubation vol.: 96 well (67 microliters / well) Function: Stable liver function until 4 weeks

Test chemicals Drug Nicotine class B Known Metabolism Routes CYP2A6, 2B6, UGT, Aldehyde oxidase etc. Dextromethorphan B CYP2D6,3A4,1A2,2C19 Imipramine Diclofenac B A CYP2D6,1A2,2C19,3A4 ; UGT1A4 CYP2C9, 3A4, UGT2B7, CL Group ml/ min / kg High CL > 15 intermediate CL 5< CL <15 In Vivo Clearance ml/ min / kg Fu CL nonrenal Ref. Ref. 18 Obach 2008 0.95 8.6 Lin, 2015 8 Lin, 2015 7.6 Lin, 2015 Obach, 2008 0.45 Witherow et al, 1999 0.72 Lutz et al, 2011 0.075 Obach, 2008 0.14 0.005 0.05 https://link.s pringer.com /article/10.1 007/BF005 68901 Obach, 2008 Dancik 2012 Tolbutamide A CYP2C9, 2C19 0.38 Lin, 2015 0.05 Obach, 2008 Low CL Warfarin A CYP2C9, CYP3A4 <5 0.05 Lin, 2015 0.015 Salicylic acid A Glycine conjugation acyl-coa N- acyltransferase, 0.21 Schwarb, 1999 0.14 0.053 Obach, 2008 Dancik 2012 https://www. ncbi.nlm.nih.gov/pubme d/9112064

HepatoPac XB170047 TS1 Tolbutamide Ln % remaining Spheroid XB160099 TS11 Tolbutamide Ln % remaining Tolbutamide Ln % remaining Suspension XB170009 TS4 Tolbutamide Ln % remaining COMPARISON SUSPENSION VS SPHEROID VS HEPATOPAC 5.5 5.0 Tolbutaminde 4.5 4.0 0 1 2 3 4 Time (hour) 5.0 4.8 4.6 4.4 4.2 4.0 0 24 48 72 96 120 144 168 Time (hour) 5.0 4.5 Suspension vs Spheroid vs HepatoPac 5 4 3 2 1 0 0 24 48 72 96 120 144 168 Time (hour) Spheroid Suspension HepatoPac 4.0 3.5 3.0 0 24 48 72 96 120 144 168 Time (hour)

Warfarin Ln % remaining SUMMARY OF CLEARANCE RATES SPHEROID VS HEPATOPAC Spheroid Test article Mean Cl int (µl/min/10 6 cells) Goodness of fit dextromethorphan 1.3 0.02 diclofenac 21.2 0.98 imipramine 14.9 0.77 salicylic acid 0.8 0.01 S-nicotine 4.5 0.85 tolbutamide 3.3 0.40 warfarin 0.8 0.01 Diclofenac HepatoPac Test article Mean Cl int 4 (µl/min/10 6 cells) Spheroid Goodness of fit Dextromethorphan 15.72 3 0.93 5 Diclofenac 2 13.21 0.69 Imipramine 9.30 1 0.99 salicylic acid 2.56 0 0.97 S-nicotine 5.35 Time (hour) 0.99 tolbutamide 2.34 1/slope 43790.97 warfarin 1.19 R square 0.01444 0.99 0 24 48 72 96 120 144 168

METABOLITE IDENTIFICATION Salicylic acid metabolites [nmol] salicylic acid metabolites in media [nmol] salicylic acid metabolites in lysate [nmol] Salicylic acid metabolite formations HepatoPac cultures 0.008 Spheroid 0.025 HepatoPac [media] 0.0008 HepatoPac [lysate] 0.006 0.020 0.0006 0.004 0.002 0.015 0.010 0.005 0.0004 0.0002 0.000 0 24 48 72 96 120 144 168 Time (hour) Hydroxyhippuric Acid Salicylic Acid Acyl Glucuronid Salicylic Acid Phenyl Glucuron Dihydroxybenzoic Acid 0.000 0 24 48 72 96 120 144 168 Time (hour) O-Hydroxyhippuric acid Salicylic Acid Acyl Glucuronid Salicylic Acid Phenyl Glucuron SAAG-stromal** ** SAAG was detected in only media of Stromal cells * No detection of 2,5-dihydroxybenzoic acid 0.0000 0 24 48 72 96 120 144 168 Time (hour) O-Hydroxyhippuric acid Salicylic Acid Acyl Glucuronid Salicylic Acid Phenyl Glucuron SAAG-stromal** ** SAAG was detected in only media of Stromal cells * No detection of 2,5-dihydroxybenzoic acid

Salicylic acid metabolites [nmol] METABOLITE IDENTIFICATION Salicylic acid metabolite formation HepatoPac 0.030 0.025 0.020 0.015 0.010 0.005 0.000 0 24 48 72 144 168 Time (hour) SAAG SAPG O-Hydroxyhippuric acid SAAG SAPG O-Hydroxyhippuric acid, lysate SAAG, lysate SAPG, lysate * No detection of 2,5-dihydroxybenzoic acid

NEXT STEPS Evaluation of PBK models Increase numbers of chemicals in evaluation of systemic predictions using other dosing routes Determine uncertainty in skin penetration studies using alternative literature data Hepatic metabolism Donor cells used from HepatoPac compared with suspension Metabolite identification from HepatoPac More donors and chemicals in HepatoPac Additional low clearance compounds HepatoPac vs. suspension Measure transcriptional changes in HepatoPac assay

ACKNOWLEDGEMENTS Unilever Richard Cubberley Mi-Young Lee Hequn Li David Sheffield Beate Nicol Joe Reynolds Ruth Pendlington Juliette Pickles Solvo Biotechnology