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

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

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

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

Evaluation of Drug-Drug Interactions FDA Perspective

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

Drug Interactions, from bench to bedside

Supplemental material to this article can be found at:

A unified in vivomodelingapproach for quantitative prediction of the impact of gene polymorphism and drug interactions on drug exposure

Considerations from the IQ Induction Working Group in Response to Drug-Drug Interaction

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

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

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

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

CYP3A Induction Can Predict P-gp Induction: An Example of Sofosbuvir (a P-gp Substrate) with Rifampin, Carbamazepine or Rifabutin

MODELING MECHANISM BASED INACTIVATION USING PBPK JAN WAHLSTROM DIRECTOR, PRECLINICAL

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

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

Supplemental material to this article can be found at:

FDA s Clinical Drug Interaction Studies Guidance (2017 Draft Guidance)

A 96-well Plate Assay for CYP3A Induction Using Cryopreserved Human Hepatocytes. Naomi Kamiguchi, Eiji Aoyama, Teruaki Okuda, and Toshiya Moriwaki

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

Supplemental material to this article can be found at:

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

Constitutive Regulation of P450s by Endocrine Factors

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

DMD Comparison of Different Algorithms for Predicting Clinical Drug-Drug. Interactions, Based on the Use of CYP3A4 in Vitro Data;

The Application of Physiologically Based Pharmacokinetic Modeling to Predict the Role of Drug Transporters: Scientific and Regulatory Perspectives

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

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

Prediction of in vivo hepatic clearance and DDI of OATP substrates: Comparison of different in vitro approaches. Yuichi Sugiyama

PBPK modeling of renal impairment what is missing?

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

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

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

Mechanisms and Predictions of Drug-Drug Interactions of the Hepatitis C Virus 3-Direct Acting

Exploiting BDDCS and the Role of Transporters

Welcome to the webinar... We will begin shortly

Hepatic Transporter Proteins involved in Bile Formation

Using virtual populations to solve drug development problems. Iain Gardner Head of Translational DMPK Science SIMCYP

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

When does the rate-determining step in the hepatic clearance of a drug switch from

Critical review of the literature on drug interactions

Modeling, Prediction, and In Vitro In Vivo Correlation of CYP3A4 Induction

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

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

Examining the Basis of Drug-Drug Interaction (DDI) Labeling Recommendations for Antiviral Approvals from 1998 to 2015

Cryo Characterization Report (CCR)

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

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

Brainteaser NK-1 receptor antagonists

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

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

Simulation and Prediction of the Drug-Drug Interaction Potential of Naloxegol by Physiologically Based Pharmacokinetic Modeling

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

Application of Physiologically Based Pharmacokinetic Modeling in. Understanding Bosutinib Drug-Drug Interactions: Importance of Intestinal

Leslie Z. Benet, PhD. Professor of Bioengineering and Therapeutic Sciences Schools of Pharmacy and Medicine University of California San Francisco

HTPK: Conducting PK modeling and

Prediction of CYP3A-Mediated Drug-Drug Interactions Using Human Hepatocytes Suspended in Human Plasma S

Supplemental Information

Inhibition of Human Hepatic Bile Acid Transporters as Contributing Factors to Drug-Induced Liver Injury

Model-Informed Drug Development (MIDD) for Ixazomib, an Oral Proteasome Inhibitor

Dose Response Approaches for Nuclear Receptor Mediated Modes of Action Workshop Preliminary Report

3. P450 Drug Metabolism DDIs: Induction

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

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

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

Evaluation of P450 Inhibition and Induction by Artemisinin Antimalarials in Human Liver Microsomes and Primary Human Hepatocytes

Physiologically Based Pharmacokinetic Model to Assess the Influence of Blinatumomab-Mediated Cytokine Elevations on Cytochrome P450 Enzyme Activity

Application of a Physiologically Based Pharmacokinetic Model to Predict OATP1B1-Related Variability in Pharmacodynamics of Rosuvastatin

Stereoselective inhibition of CYP2C19 and CYP3A4 by fluoxetine and its metabolite:

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

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

Pharmacologic Considerations when using DAAs in Cirrhosis

Pursuing the holy grail of predicting and verifying tissue drug concentrations: A proteomics and PET imaging approach

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

Evaluation of clinical drug interaction potential of clofazimine using static and dynamic

Application of PBPK Modeling and Simulations in Drug Development

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

Transporter-mediated disposition, clinical pharmacokinetics and cholestatic potential of glyburide and its primary active metabolites

Examination of the Human Cytochrome P4503A4 Induction Potential of PF , an Irreversible Myeloperoxidase Inactivator: Integration of

Combination therapy with simeprevir and TMC647055/low dose ritonavir: dose anticipation using PBPK modeling and dose optimization in healthy subjects

Identification of influential proteins in the classical retinoic acid signaling pathway

Physiologically Based Pharmacokinetic Model of Mechanism-Based Inhibition of CYP3A by Clarithromycin

Effects of Uptake and Efflux Transporter Inhibition on Erythromycin Breath Test Results

Opinion on. Classification of Musk ketone

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

Supplemental material to this article can be found at:

A study on pharmacokinetics of bosentan with systems modeling, Part 1: translating systemic plasma concentration to liver exposure in healthy subjects

Hepatic Drug-Metabolizing Enzyme Induction and Implications for Preclinical and Clinical Risk Assessment

Muhammad Fawad Rasool Feras Khalil Stephanie Läer

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

Role of CYP3A4 in kinase inhibitor metabolism and assessment of CYP3A4 activity

Pharmacokinetic drug drug interactions of tyrosine kinase inhibitors: A focus on cytochrome P450, transporters, and acid suppression therapy

Evaluation of the Interplay between Uptake Transport and CYP3A4 Induction in Micropatterned Cocultured Hepatocytes s

Simulation of Clinical Drug-Drug Interactions from Hepatocyte CYP3A4 Induction Data and Its Potential Utility in Trial Designs

Proteomic Quantification of Kidney Transporters: Methodological Challenges, Interindividual Variability and Application in IVIVE

Use of Immortalized Human Hepatocytes to Predict the Magnitude of Clinical. Drug-Drug Interactions Caused by CYP3A4 Induction ξ

An Evaluation of the Dilution Method for Identifying Metabolism-Dependent Inhibitors of Cytochrome P450 Enzymes

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

Hepatic Efflux Transporters: Relevance to Drug-Drug Interactions and Drug Toxicity

Transcription:

The Future of In Vitro Systems for the Assessment of Induction and Suppression of Enzymes and Transporters AAPS, San Diego November 6 th, 2014 Andrew Parkinson, XPD Consulting Lisa Almond, Simcyp-Certara Jane Kenny, Genentech Harma Ellens, GSK 1

Induction and Suppression Induction Can increase clearance and cause loss of therapeutic action (e.g., OCS) Xenosensor FDA CYP Control Inducer AhR CYP1A2 Omeprazole Direct agonist CITCO Direct agonist CAR CYP2B6 Phenobarbital Indirect agonist PXR CYP3A4 Rifampin Direct agonist Suppression Can decrease clearance Activators of other nuclear receptors Cytokines (e.g., TNF, IL-6) NF- B activation xenosensor inactivation (and vice versa) Biologics may act directly on hepatocytes/kupffer cells or release cytokines from PBMC Inducers tend to be small drug molecules Suppressors tend to be biologics (therapeutic proteins) but there are exceptions 2

3 Induction studies before and after the FDA 2012 DDI Guidance 3 Induction assay Before 2012 After 2012 Endpoints Positive attribute Negative attribute CYP activity Fold induction Percent of positive control Endpoint = net effect of induction & irreversible inhibition CYP inhibition could mask induction CYP mrna EC 50 and E max Endpoint = induction only (inhibition examined separately) Assumes induction is always due to increased transcription (not so for CYP2C19 or 2E1) control criteria > 2 fold > 6 fold Test article (TA) concentration Up to10 x plasma C max In vitro E max TA treatment 3 days 2 or 3 days IVIVE >2 fold (or >1.5 fold) >40% (or >20%) control R 3 < 0.9

CYP3A4 induction (E max ) : mrna versus activity Predictive utility of in vitro rifampin induction data generated in fresh and cryopreserved human hepatocytes, Fa2N-4, and HepaRG cells. Templeton IE, Houston JB, Galetin A. Drug Metab Dispos 39: 1921-1929, 2011 Relatively large Relatively small 4

CYP3A4 induction (EC 50 ) : mrna versus activity Predictive utility of in vitro rifampin induction data generated in fresh and cryopreserved human hepatocytes, Fa2N-4, and HepaRG cells. Templeton IE, Houston JB, Galetin A. Drug Metab Dispos 39: 1921-1929, 2011 Comparable but Fa2N-4 EC 50 values are high 5

CYP3A4 mrna induction: EC 50 in different cells Comparison of immortalized Fa2N-4 cells and human hepatocytes as in vitro models for cytochrome P450 induction. Hariparsad N, Carr BA, Evers R, Chu X. Drug Metab Dispos 36: 1046-1055, 2008 Fa2N-4 cells EC 50 = 8 µm Human hepatocytes EC 50 = 0.8 µm 6 Levels of PXR mrna in Fa2N-4 cells human hepatocytes but CAR mrna is <1% Rifampicin is an OATP substrate Levels of OATP1B1 and OAPT1B3 mrna levels in Fa2N-4 cells <1% of hepatocytes

[Drug] Measuring the in vitro concentration of the inducing drug seems like a good idea but... where is the drug? Whole-system loss In many cases most of the drug moves from the medium to the cells after only a few minutes Medium loss 0 5 10 15 20 Time (min) 7

Measuring the in vitro concentration of the inducing drug seems like a good idea but... where is the drug? For certain basic drugs, they undergo extensive partitioning into phospholipid membranes (not shown) or ion-partitioning into the acidic lysosomes Total cell concentration >> unbound intracellular concentration 8

Measuring the in vitro concentration of the inducing drug seems like a good idea but... where is the drug? 9 For certain acidic/zwitterion drugs, they undergo transporter-mediated uptake and efflux into the bile canaliculi Total cell concentration >> unbound intracellular concentration

Should we correct for variation in EC 50? 10 Perfect results in all 3 human hepatocyte preparations. Correction? None

Should we correct for variation in EC 50? 11 A high EC 50 value observed for both the positive control and test article in 1 of 3 preparations of human hepatocyte preparations. Correction? Probably YES

Should we correct for variation in EC 50? 12 A high EC 50 value observed for ONLY the positive control in 1 of 3 preparations of human hepatocyte preparations. Correction? Case-by-case

In vitro to in vivo extrapolation (IVIVE) A comparison of 5 models of CYP3A4 induction Evaluation of various static and dynamic modeling methods to predict clinical CYP3A induction using in vitro CYP3A4 mrna induction data. Einolf HJ, Chen L, Fahmi OA, Gibson CR, Obach RS, Shebley M, Silva J, Sinz MW, Unadkat JD, Zhang L, Zhao P. Clin Pharmacol Ther 95: 179-188, 2014 13

In vitro to in vivo extrapolation (IVIVE) A comparison of 5 models of CYP3A4 induction Correlation approach = C max EC 50 Relative induction score Basic static model (R 3 ) 14

In vitro to in vivo extrapolation (IVIVE) A comparison of 5 models of CYP3A4 induction Mechanistic static model (AUC Ratio) Liver Intestine where A, B and C are terms for TDI, induction, and reversible inhibition in the liver, respectively and X, Y and Z are terms for TDI, induction, and reversible inhibition in the intestine, respectively Mechanistic dynamic model (modeling & simulation) 15 AUCR + PBPK modeling. Incorporates such variables such as time-dependent changes in inducer concentration, which is complex when the drug is an auto-inducer

In vitro to in vivo extrapolation (IVIVE) A comparison of 5 models of CYP3A4 induction Model False negative False positive True negative True positive Within 2 fold Correlation 0 3 3 22 23 RIS 0 2 4 22 19 R 3 (d = 1) 0 3 3 22 17 Mechanistic Static 2 (0) 0 (3) 6 (3) 20 (22) 19 (20) Mechanistic Dynamic 2 (0) 1 (3) 5 (3) 20 (22) 11 (14) 16 [I] = total plasma C max (bound + unbound) Values in parentheses are without the time-dependent inhibition component, which tends to be overestimated for CYP3A4

The FDA s R 3 Raising the negative result bar If we assume d=1 and use plasma C max as the in vivo inducer concentration, then the equation for R 3 is: 1 R 3 = 1 + E max + C max EC 50 + C max The bare minimum cutoff value of is R 3 is 0.8999 (marginally below 0.9). This value is obtained when: E max + C max EC 50 + C max = 0.1112 Accordingly, EC 50 C max = E max 0.1112 1 To fall just below the FDA s cutoff of 0.9, EC 50 (as a multiple of C max ) increases almost linearly with E max. 17

The FDA s R 3 Raising the negative result bar This graph shows the relationship between EC 50 (as a multiple of C max ) and E max when the value of R 3 is 0.899 (marginally below the FDA s cutoff of 0.9) Note the near linear relationship General Rule You are safe if EC 50 is greater than C max x 10 E max 18

The FDA s R 3 Raising the negative result bar This graph shows the dividing line between a positive and negative result for CYP induction when E max = 10 (the induced activity was 11 times control) Note that the EC 50 value is 89 times C max 19

The FDA s R 3 Raising the negative result bar This graph shows a series positive results for CYP induction when E max = 10. R 3 values range from 0.9 (safe) to 0.167, which is achieved when EC 50 = C max. Note that when the in vitro concentration of inducer equals C max (or even 10 x C max ), the degree of induction is far less than 40% of E max in the majority of cases 20

The FDA s R 3 Raising the negative result bar This graph shows the dividing line between a positive and negative result for CYP induction when E max ranges from 1 to 10 (the induced activity was 2 to 11 times control) Note that for ALL these safe curves, negligible induction is observed when the in vitro concentration of inducer equals C max regardless of the value of E max 21

The FDA s R 3 Raising the negative result bar To be sure of a negative result (defined as an R 3 of 0.9 or greater), it is necessary to test concentrations of test article that exceed C max As E max increases (as it does when mrna levels are measured) so must the concentration of test article (as a multiple of C max ) to demonstrate no potential for clinical induction 22