DRUG METABOLISM AND PHARMACOKINETICS (DMPK) Lena Gustavsson, H. Lundbeck A/S, November 2015

Similar documents
Exploiting BDDCS and the Role of Transporters

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

Drug Interactions, from bench to bedside

Building innovative drug discovery alliances. Hepatic uptake and drug disposition o in vitro and in silico approaches

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

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

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

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

Pharmacokinetic Modeling & Simulation in Discovery and non-clinical Development

Biopharmaceutics Drug Disposition Classification System (BDDCS) and Its Application in Drug Discovery and Development

Evaluation of Drug-Drug Interactions FDA Perspective

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

Biopharmaceutics Drug Disposition Classification System (BDDCS) --- Its Impact and Application

T Eley, Y-H Han, S-P Huang, B He, W Li, W Bedford, M Stonier, D Gardiner, K Sims, P Balimane, D Rodrigues, RJ Bertz

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

Physiologically-Based Simulation of Daclatasvir Pharmacokinetics With Antiretroviral Inducers and Inhibitors of Cytochrome P450 and Drug Transporters

EVALUATION OF DRUG-DRUG INTERACTION POTENTIAL BETWEEN SACUBITRIL/VALSARTAN (LCZ696) AND STATINS USING A PHYSIOLOGICALLY- BASED PHARMACOKINETIC MODEL

MODULE PHARMACOKINETICS WRITTEN SUMMARY

Pharmacogenetics and Pharmacokinetics

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

Constitutive Regulation of P450s by Endocrine Factors

Cytochrome P450 Drug Interaction Table Flockhart Table

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

Transporters DDI-2018

Efficient Liver Targeting and Uptake by Novel Tenofovir Prodrug, CMX157, For the Treatment of Hepatitis B

Welcome to the webinar... We will begin shortly

Itraconazole and Clarithromycin as Ketoconazole Alternatives for Clinical CYP3A Inhibition Studies to Quantify Victim DDI Potential

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

Lecture 8: Phase 1 Metabolism

1 Introduction: The Why and How of Drug Bioavailability Research

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

What Can Be Learned from Recent New Drug Applications? A Systematic Review of Drug

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

Cytokrom P450 (CYP) Hepatic Drug Metabolism. Medicines in plasma. Plasma concentration of a medicine. Eva Brittebo Dept Pharmaceutical Biosciences

Evaluation of Proposed In Vivo Probe Substrates and Inhibitors for Phenotyping Transporter Activity in Humans

Erik Mogalian, Polina German, Chris Yang, Lisa Moorehead, Diana Brainard, John McNally, Jennifer Cuvin, Anita Mathias

DRUG ELIMINATION II BILIARY EXCRETION MAMMARY, SALIVARY AND PULMONARY EXCRETION

Click to edit Master title style

DRUG-DRUG INTERACTIONS OF GLECAPREVIR AND PIBRENTASVIR WITH PRAVASTATIN, ROSUVASTATIN, OR DABIGATRAN ETEXILATE

3. P450 Drug Metabolism DDIs: Induction

WHY... 8/21/2013 LEARNING OUTCOMES PHARMACOKINETICS I. A Absorption. D Distribution DEFINITION ADME AND THERAPEUIC ACTION

Using Accelerator Mass Spectrometry to Explain the Pharmacokinetics of Vismodegib Cornelis E.C.A. Hop

Practical Application of PBPK in Neonates and Infants, Including Case Studies

Regulation of the cell surface expression and transport capacity of BSEP by small chemical molecules

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

BASIC PHARMACOKINETICS

Principles of Toxicokinetics/Toxicodynanics

Pharmacokinetics in Drug Development. Edward P. Acosta, PharmD Professor & Director Division of Clinical Pharmacology Director, CCC PK/PD Core

Cryo Characterization Report (CCR)

In vitro substrate-dependent inhibition of OATP1B1 and its impact on DDI prediction

RISK FACTORS AND DRUG TO STATIN-INDUCED MYOPATHY

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

Complexities of Hepatic Drug Transport: How Do We Sort It All Out?

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

Strategies for Developing and Validating PBPK Models for Extrapolation to Unstudied Population

The Influence of Physicochemical Properties on ADME

Core Data Set CYP2D6 Metabolism

HUMAN BIOTRANSFORMATION. ANDREW PARKINSON, BRIAN W. OGILVIE, BRANDY L. PARIS, TIFFINI N. HENSLEY, and GREG J. LOEWEN XenoTech LLC, Lenexa, KS

Understand the physiological determinants of extent and rate of absorption

Effects of Liver Disease on Pharmacokinetics

Introduction to and Application of Pharmacokinetics in Pharma R and D

Investigating Transporter-Mediated Drug-Drug Interactions Using a Physiologically Based Pharmacokinetic Model of Rosuvastatin

Pharmacokinetics: The Basics

MODELING MECHANISM BASED INACTIVATION USING PBPK JAN WAHLSTROM DIRECTOR, PRECLINICAL

Supplemental Information

Effect of Multiple-Dose Ketoconazole and the Effect of Multiple-Dose Rifampin on Pharmacokinetics (PK) of the HCV NS3 Protease Inhibitor Asunaprevir

We will begin momentarily at 2pm ET. Slides available now! Recordings will be available to ACS members after one week.

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

Prediction of the Effects of Renal Impairment on the Clearance for Organic Cation Drugs that. undergo Renal Secretion: A Simulation-Based Study

Critical review of the literature on drug interactions

The extended clearance model and its use for the interpretation of hepatobiliary elimination data

Supplemental Materials

Use of PBPK in simulating drug concentrations in pediatric populations: Case studies of Midazolam and Gabapentin

BIOPHARMACEUTICS and CLINICAL PHARMACY

Pharmacogenetics of Codeine. Lily Mulugeta, Pharm.D Office of Clinical Pharmacology Pediatric Group FDA

Evaluation and Quantitative Prediction of Renal Transporter-Mediated Drug-Drug Interactions. Bo Feng, Ph.D. DDI 2017 June 19-21, 2017

Muhammad Fawad Rasool Feras Khalil Stephanie Läer

Case #1. Pharmacology and Drug Interactions of Newer Direct-Acting Antivirals

FDA Use of Big Data in Modeling and Simulations

The ADME properties of most drugs strongly depends on the ability of the drug to pass through membranes via simple diffusion.

UNIVERSITY OF THE WEST INDIES, ST AUGUSTINE

Section 5.2: Pharmacokinetic properties

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

HTPK: Conducting PK modeling and

CO-ADMINISTRATION WITH GRAZOPREVIR AND ELBASVIR HAS NO EFFECT ON PRAVASTATIN EXPOSURE BUT INCREASES ROSUVASTATIN EXPOSURE IN HEALTHY SUBJECTS

USING PBPK MODELING TO SIMULATE THE DISPOSITION OF CANAGLIFLOZIN

Development of Canagliflozin: Mechanistic Absorption Modeling During Late-Stage Formulation and Process Optimization

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

Metformin: Mechanistic Absorption Modeling and IVIVC Development

Physiologically Based Pharmacokinetic Model of the CYP2D6 Probe Atomoxetine: Extrapolation to Special Populations and Drug-Drug Interactions s

Falk Symposium 156: Genetics in Liver Disease. Pharmacogenetics. Gerd Kullak-Ublick

DARUNAVIR/COBICISTAT MODULE PHARMACOKINETICS WRITTEN SUMMARY

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

Biopharmaceutics. Tips Worth Tweeting. Contributor: Sandra Earle

Basic Biopharmaceutics, Pharmacokinetics, and Pharmacodynamics

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

Pharmacokinetic and absolute bioavailability studies in early clinical development using microdose and microtracer approaches.

Clinical Pharmacokinetics of Tyrosine Kinase Inhibitors

Current and Emerging Transporter Regulatory Themes in Drug Development: Relevance to Understanding PK/PD, DDIs, and Toxicity

Clinical Pharmacology of DAA s for HCV: What s New and What s in the Pipeline

Transcription:

DRUG METABOLISM AND PHARMACOKINETICS (DMPK), H. Lundbeck A/S, LEGU@lundbeck.com November 2015

DMPK in Drug Discovery and Development Agenda Introduction Optimizing pharmacokinetic properties Absorption & bioavailability Distribution Elimination Clearance Understanding clearance mechanisms Drug metabolism Drug transporters Drug drug interactions and interindividual variability Summary: Drug discovery and development 2

H. Lundbeck A/S an introduction A pharmaceutical company with focus on brain diseases More than 700 million people are affected by brain disease worldwide Lundbeck is dedicated to address the global burden of brain disease Psychiatric diseases e.g. bipolar disorder, depression, schizophrenia Neurologic diseases e.g. Alzheimer s, Parkinson s, Huntington s A global company with head quaters in Valby, Denmark Total approximately 5500 employee s Approximately 1700 employees in Denmark Full value chain from research to production Want to know more? Go to: www.lundbeck.com/global/about-us/progress-in-mind www.youtube.com/user/progressinmind 3

Why study Drug Metabolism and PharmacoKinetics? DRUG DISCOVERY Optimise compounds to get... Good bioavailability get to its target Appropriate duration (1-2 doses/day) Low potential for drug-drug interactions predicted to man Provide basics for understanding of Toxicology Pharmacology From Rowland and Tozer, 1995 DRUG DEVELOPMENT Provide understanding of drug disposition Preclinical animal species tox coverage Human data Assess the risk for drug drug interactions (DDI) Decrease risk for drug drug interactions in the clinic Impact on the design of clinical studies Comply with guidelines from regulatory authorities

Reasons for compound attrition Kola and Landis 2004

Pharmacokinetics oral administration Drug concentration in plasma C max t max ADME = Absorption Distribution Metabolism Excretion

DMPK in Drug Discovery and Development Agenda Introduction Optimizing pharmacokinetic properties Absorption & bioavailability Distribution Elimination Clearance Understanding clearance mechanisms Drug metabolism Drug transporters Drug drug interactions and interindividual variability Summary: Drug discovery and development 7

Absorption Lipinski s rule of 5 to predict poor permeability/absorption (Lipinski et al, Adv Drug Delivery Rev 23:3-25, 1997) Mw > 500 Log P > 5 H-bond donors >5 H-bond acceptors > 10 Transporter substrates are exceptions from the rule.

Permeability Caco-2 Is the drug absorbed? No = low bioavailability! Papp: cm/sec x 10-6 correlates to human abs Indication of transporter mechanisms Automated incubations LC-MS/MS analysis Caco-2 Human colon epithelial cell line Differentiates to monolayer with tight junctions Alternative to Caco-2: PAMPA artificial membrane

Bioavailability - oral administration F = F gut x F abs x F hep Gut lumen Gut wall F gut F abs Portal vein Liver F hep F F hep = 1 - E hep

Distribution Drug distribution is the reversible transfer of drug to and from the site of measurement (blood/plasma) Distribution is influenced by -perfusion blood circulation to tissues -diffusion -physicochemical properties -binding to proteins etc From Rowland and Tozer, 1995

Volume of distribution (V) Not a real volume but a mathematical expression of the extent to which a drug distributes into tissues -Low V drug stays in blood/plasma -High V drug distributes extensively into tissues V relates the concentration at site of measurement to the total amount of drug in the body (L/kg) V=Amount drug in body/plasma concentration (L/kg bw)

Elimination: The concept of clearance (CL) Clearance is the apparent volume of plasma completely cleared of drug per unit time Rate of elimination = CL x C CL = Dose / AUC (iv dose) Unit: ml/min/kg

Hepatic clearance Hepatic vein Gall Bladder Bile duct Hepatic portal vein Hepatic artery The liver is the major site of drug metabolism

Drug metabolizing enzymes Route of elimination of the top 200 most prescribed drugs in 2002 Enzymes listed in FDA guidelines CYP: 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 3A UGT: 1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15 Weinkers and Health Nat Rev Drug Discov 4:825-833, 2005

How to estimate metabolic clearance from in vitro studies?

Phase I +II II Metabolic stability Microsomes or hepatocytes How fast is the drug eliminated by the liver? Fast = low bioavailability! Fast = short duration! CL int - the intrinsic capacity of a system to clear a drug (µl/min/mg protein or cells) metabolites CL int = V max K m = V 0 / [S] ln Substrate Concentration 100 10 1 0 20 40 60 80 0,1 0,01 Time (minutes)

Prediction of in vivo clearance from in vitro data In vitro t 1/2 CL int = ln 2 /( t 1/2 x protein conc) In vitro CL int CL int = CL int x (mg microsomes/g liver) x (g liver/kg bw) Whole liver CL int Hepatic metabolic CL Whole body CL If well-stirred model CL hep,met, = (Q h x f u x CL int )/ (Q h + (f u x CL int ) CL = CL(HepMet)+CL(HepBile)+CL(Renal)+...

Interplay between V and CL Rat pharmacokinetics 10000 Concentration (nmol/l) 1000 100 10 C B A Elimination half-life T 1/2 = ln 2 x V / CL 1 0 5 10 15 Time (hours) CL(mL/min/kg) Vss(L/kg) T½ (h) A 20 14 10 B 70 12 3 C 80 0.6 0.5

Volume of distribution Clearance Absorption Half-life Oral bioavailability Dosing interval? Dose?

DMPK in Drug Discovery and Development Agenda Introduction Optimizing pharmacokinetic properties Absorption & bioavailability Distribution Elimination Clearance Understanding clearance mechanisms Drug metabolism Drug transporters Drug drug interactions and interindividual variability Summary: Drug discovery and development 21

Clearance mechanisms Total CL = CLMetHep + CLMetBile + CLRenal +... Hepatic Metabolism phase I and phase II enzymes Bile excretion sinusoidal and canalicular transporters Renal Passive glomerular filtration Active transport Extrahepatic metabolism Intestinal CYP3A4 Enzymes in blood Other extrahepatic enzymes

Hepatic clearance mechanisms Sinusoidal membrane Blood Drug Hepatocyte Drug Drug uptake transporters Metabolite Drug metabolising enzymes Canalicular membrane Bile canaliculus Efflux transporters

Uptake & Efflux Transporters SLCs Solute Carriers o OAT Organic Anion Transporter o OCT Organic Cation Transporter o OATP Organic Anion Transporting Polypeptides ABC series ATP Binding Cassette transporters o MDR Multi Drug Resistance proteins o MRP Multi drug Resistance-like Proteins o White family Drosophila white eye pigment gene Blood Hepatocyte

Drug transporters that influences drug disposition clinical evidence From International Transporter Consortium Giacomini et al Nature Rev Drug Disc 2010 Modified marking EMA recommended transporters in blue

Pravastatin O O O O O H OChiral 3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor; key enzyme in cholesterol synthesis Used for the management of hypercholesterolaemia The target is in the liver Has short t 1/2 (~2h), low F (17%) but successful Has a good safety profile compared to other statins WHY? O

Disposition of Pravastatin Oral Oral tablet tablet Substrate for OATP1B1 MRP2 Gut OATP1B1 Liver MRP2 Enterohepatic recirculation t of t Systemic Circulation Active secretion Kidney

Simvastatin-induced myopathy increased due to increased plasma exposure - OATP1B1 polymorphism Niemi, Clin.Pharm.Ther. 2010 Pasanen et al, Pharmacogenet. Genomics 2006 Search study N.Engl.J.Med. 2008

Prediction of Human PK In vitro, human Species differences Scaling CL Absorption (Caco-2) Drug-drug interactions In vitro, animal In vivo, human In vitro/in vivo correlation Allometric scaling Vss (dog, human PPB) Absorption (rat) In vivo, animal

DMPK in Drug Discovery and Development Agenda Introduction Optimizing pharmacokinetic properties Absorption & bioavailability Distribution Elimination Clearance Understanding clearance mechanisms Drug metabolism Drug transporters Drug drug interactions and interindividual variability Summary: Drug discovery and development 30

Interindividual variability Age Sex Genetics Enzyme content Liver weight Organ blood flow... Nature Reviews Drug Discovery 6, 140-148 (February 2007) doi:10.1038/nrd2173 http://www.simcyp.com

Interindividual variation in drug response

CYP2D6 phenotypes in a Swedish population

Codeine metabolism to morphine is metabolised by CYP2D6 CYP2D6 CYP2D6 Poor metabolizer Codeine Prodrug No formation of morphine Morphine Active metabolite Lack of analgesia CYP2D6 Ultra-rapid metabolizer Formation of morphine Overdosing Adverse events

Drug drug interactions - CYP inhibition Does the drug inhibit Cytochrome P450? Yes = Potential drug interactions! P450 P450 metabolite metabolite

Metabolism of terfenadine OH OH CYP3A4 OH OH N N COOH Terfenadine Almost complete first pass extraction in man Active Metabolite Responsible for efficacy in man

Ketoconazole Ketoconazole is an antifungal agent O N N O O Cl O N N Potent inhibitor of CYP3A4 IC 50 value <1µM Antifungal dose is high (400mg twice daily) Circulating concentrations of ketoconazole exceed IC 50 for CYP3A4 inhibition Cl

Ketoconazole terfenadine interaction OH OH CYP3A4 OH OH N N COOH High circulating concentrations of terfenadine Low circulating concentrations of metabolite

Implications of terfenadine ketoconazole interaction High circulating concentrations of terfenadine Potential to prolong QT interval of the ECG Abnormal heart rhythm Small numbers of patients go on to develop fatal Torsade de Pointes (heart stops) Led to withdrawal of terfenadine from the market Increased questioning of Regulatory Authorities on QT and DDIs

CYP inhibition Recombinant enzymes Human liver microsomes CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4 Human recombinant P450 enzymes substrate CYP product * IC50 = µm If IC50 < 10 µm potential interaction If [I]/Ki >0.1 - need to address in clinical study Discovery: Automated fluorescence based Development: LC-MS/MS analysis of metabolite

Induction of P450 enzymes Transcriptional regulation by nuclear hormone receptors Aryl hydrocarbon Receptor (AhR) Ligands: Polyaromatic hydrocarbons, dioxins (TCDD), Omeprazol Target genes: CYP1A1, CYP1A2, CYP1B1 Arnt - NUCLEUS - Constitutive Androstane Receptor (CAR) Ligands: Phenobarbital, CITCO Target genes: CYP2B6 Pregnane X Receptor (PXR) Ligands: Rifampin, Carbamazepine Target genes: CYP3A4, CYP2C8, CYP2C9, CYP2C19 PAH PB Rif AhR Hsp90 CAR PXR, GR? RXR RXR RXR CYP1A CYP2B CYP3A CYP2C Cross-talk between nuclear hormone receptors (AhR, CAR, PXR, GR, Hnf4 etc)

DDI Risk Assessment Victim (substrate) Enzyme/transporter phenotyping Drug disposition e.g. clearance Fraction of total elimination Mechanistic understanding Perpetrator (inhibitor/inducer) Enzyme/transporter IC 50 /K i Concentration plasma, liver, intestine Bound vs unbound Time dependence Also includes polymorphism Complex interactions how to assess the risk? Integration of data my modeling and simulation PBPK Iterative addition of new data Other relevant information - Co-medications - Biopharmaceutical Classification System etc

Physiology Based Pharmacokinetic (PBPK) Modelling and Simulation Jones and Rowland-Yeo 2013

PBPK modelling and simulation A DDI example compound A Compound A is mainly metabolized by CYP3A4 Assessment of DDI risks with compound A as a victim How will the plasma concentration change when codosing a potent CYP3A4 inhibitor How will the plasma concentration curve change when co-dosing with a strong inducer of CYP3A4? Median % fm and fe in absence of inhibitor(s) CYP3A4 Liver CYP3A5 Liver Renal

Prediction of the effect of a CYP3A4 inhibitor on the AUC of compound A 250E+00 Systemic Concentration (ng/ml) 200E+00 150E+00 100E+00 050E+00 200 mg itraconazole QD x 20 days 3 mg compound A on day 12 AUC ratio = 3.1 0.000E+00 0 45 90 135 180 225 270 315 360 405 450 Time - Substrate (h) CSys CSys with Interaction Co-administration of itraconazole (potent CYP3A4 inhibitor) may result in a 3 fold increase in the AUC of compound A A clinical DDI study is required to investigate the effect in vivo 45

Simulation of plasma concentration curves prediction of the effect of a CYP3A4 inducer Day 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 Compound AF34134 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 A rif 600 600 600 600 600 600 600 600 600 600 600 Compound A Co-administration of a strong CYP3A4 inducer, rifampicin, with compound A leads to a decrease in AUC to 20% High risk of loosing the pharmacological efficacy of compound A Perform a clinical study to assess risk in vivo

DMPK in Drug Discovery and Development Agenda Introduction Optimizing pharmacokinetic properties Absorption & bioavailability Distribution Elimination Clearance Understanding clearance mechanisms Drug metabolism Drug transporters Drug drug interactions and interindividual variability Summary: Drug discovery and development 47

Understanding and predicting drug disposition an iterative process of data integration Input data

Why study Drug Metabolism and PharmacoKinetics? DRUG DISCOVERY Optimise compounds to get... Good bioavailability get to its target Appropriate duration (1-2 doses/day) Low potential for drug-drug interactions predicted to man Provide basics for understanding of Toxicology Pharmacology From Rowland and Tozer, 1995 DRUG DEVELOPMENT Provide understanding of drug disposition Preclinical animal species tox coverage Human data Assess the risk for drug drug interactions (DDI) Decrease risk for drug drug interactions in the clinic Impact on the design of clinical studies Comply with guidelines from regulatory authorities

50 THANKS FOR LISTENING!