PBPK modeling of renal impairment what is missing?

Similar documents
Evaluation of Drug-Drug Interactions FDA Perspective

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

FDA s Clinical Drug Interaction Studies Guidance (2017 Draft Guidance)

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

Application of PBPK Modeling and Simulations in Drug Development

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

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

PHYSIOLOGICAL SCALING FACTORS AND MECHANISTIC MODELS FOR PREDICTION OF RENAL CLEARANCE FROM IN VITRO DATA

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

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

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

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

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

Sub Population: The elderly

Modeling and predicting drug pharmacokinetics in patients with renal impairment

Effects of Renal Disease on Pharmacokinetics October 8, 2015 Juan J. L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program

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

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

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

Development of a Dynamic Physiologically Based Mechanistic Kidney Model to Predict Renal Clearance

Renal Disease and PK/PD. Anjay Rastogi MD PhD Division of Nephrology

MODULE PHARMACOKINETICS WRITTEN SUMMARY

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

Effects of Renal Disease on Pharmacokinetics

Detail features... 1

Exploiting BDDCS and the Role of Transporters

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

Monica Edholm Medica Medic l a Pr oducts Agency

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

Welcome to the webinar... We will begin shortly

PHA Second Exam. Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

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

Transporters DDI-2018

Muhammad Fawad Rasool Feras Khalil Stephanie Läer

Nephron Function and Urine Formation. Ms. Kula December 1, 2014 Biology 30S

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

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

organs of the urinary system

BLOCK REVIEW Renal Physiology. May 9, 2011 Koeppen & Stanton. EXAM May 12, Tubular Epithelium

MODELING MECHANISM BASED INACTIVATION USING PBPK JAN WAHLSTROM DIRECTOR, PRECLINICAL

Excretion of Drugs. Prof. Hanan Hagar Pharmacology Unit Medical College

Nephron Structure inside Kidney:

Effect of Chronic Kidney Disease on Nonrenal Elimination Pathways: A Systematic Assessment of CYP1A2, CYP2C8, CYP2C9, CYP2C19, and OATP

PHA First Exam. Fall 2004

PHA First Exam Fall 2003

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

REVISITING THE PHARMACOKINETICS IN PATIENTS

Collin College. BIOL Anatomy & Physiology. Urinary System. Summary of Glomerular Filtrate

BCH 450 Biochemistry of Specialized Tissues

Pharmacokinetic Modeling & Simulation in Discovery and non-clinical Development

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

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

Effects of Liver Disease on Pharmacokinetics

28/04/2013 LEARNING OUTCOME C13 URINARY SYSTEM STUDENT ACHIEVEMENT INDICATORS STUDENT ACHIEVEMENT INDICATORS URINARY SYSTEM & EXCRETION

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

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

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

Drug Dosing in Renal Insufficiency. Coralie Therese D. Dimacali, MD College of Medicine University of the Philippines Manila

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

Acknowledgements. National Kidney Foundation of Connecticut Mark Perazella. Co-PI Slowing the progression of chronic kidney disease to ESRD

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

Drug Interactions, from bench to bedside

Altered GI absorption in special populations: An industry perspective

Physio 12 -Summer 02 - Renal Physiology - Page 1

The Urinary S. (Chp. 10) & Excretion. What are the functions of the urinary system? Maintenance of water-salt and acidbase

Urinary System. consists of the kidneys, ureters, urinary bladder and urethra

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

Renal System Physiology

You should know the T max for any substance that you use and for PAH ; T max = mg / min

1. Anatomy / Vascularisation. 2. Urine concentration. 3. Axial heterogeneity of some segments

Chapter 23. Composition and Properties of Urine

Chapter 25: Urinary System

Urinary Physiology. Chapter 17 Outline. Kidney Function. Chapter 17

PK-UK Challenges and benefits of using PBPK to evaluate an IVIVC for drugs with nonideal solubility and/or permeability. Bath, November 2014

Human Urogenital System 26-1

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

Renal Clearance. Dr. Eman El Eter

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

The functions of the kidney:

A. Incorrect! The urinary system is involved in the regulation of blood ph. B. Correct! The urinary system is involved in the synthesis of vitamin D.

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

Mechanistic Modeling of in vitro Assays to Improve in vitro/in vivo Extrapolation

Homeostatic Regulation

The principal functions of the kidneys

Application of a Systems Approach to the Bottom-Up Assessment of Pharmacokinetics in Obese Patients

Outline Urinary System

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

RENAL SYSTEM 2 TRANSPORT PROPERTIES OF NEPHRON SEGMENTS Emma Jakoi, Ph.D.

DILI: Clinical Pharmacology Considerations for Risk Assessment

Functional morphology of kidneys Clearance

Renal Physiology - Lectures

General Anatomy of Urinary System

Salt and Water Balance and Nitrogen Excretion

PARTS OF THE URINARY SYSTEM

Drug-Disease Modeling Applied to Drug Development and Regulatory Decision Making in the Type 2 Diabetes Mellitus Arena

BIOL2030 Human A & P II -- Exam 6

Renal Excretion of Drugs

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

Drug Absorption and Bioavailability

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

Transcription:

PBPK modeling of renal impairment what is missing? Aleksandra Galetin Centre for Applied Pharmacokinetic Research, University of Manchester, UK

Outline of the presentation Physiological changes in renal impairment PBPK modelling of RI A. Nonrenally cleared drugs B. Renally eliminated drugs System data needed for mechanistic kidney models Drug-transporter interaction in renal impairment - digoxin example

Renal elimination of drugs UGT2B7, UGT1A9 Active uptake via OAT1/3, OCT2 paired with efflux transporters MRP2/4, MATEs Proximal tubule cells also express drug metabolising enzymes Reabsorption - generally passive, active reabsorption via OAT4, PEPT1/2

Integrated bottom up and top down approach for mechanistic prediction of CL R Prediction Clinical data Physiologically- based models In vivo input Learn & Confirm In vitro input System parameters Model Parameters Estimation Neuhoff S, Gaohua L, Burt H, Jamei M, Li L, Tucker G, et al. Accounting for Transporters in Renal Clearance: Towards a Mechanistic Kidney Model (Mech KiM). In: Sugiyama Y, Steffansen B, editors. Transporters in Drug Development: Springer New York; 2013. p. 155-77.

Prediction of renal tddis and nephrotoxicity UGT2B7, UGT1A9 Recent examples cidofovir, rivaroxaban, metformin, lesinurad 1-3 In vitro transporter kinetic data and certain system parameters still sparse 4 1 Hsu Clin Pharmacokinet 2014, 2 Grillo BDD 2012, 3 Burt EJPS 2016, 4 Scotcher AAPS J 2016

Where we can expect PBPK modelling to inform drug labelling in the future? Large % of drug labels for FDA approved drugs in 2013-14 lack dose recommendations in RI Jadhav J Clin Pharmacol 2015 Younis J Clin Pharmacol 2016

Changes in system parameters in CKD KIDNEY 1-5 CL R LIVER 2,6-8 Q R and kidney weight GFR Stages 1-5: 90 to <15 ml/min/1.73m 2 CL H for nonrenally cleared drugs Downregulation or inhibition of CYPs activity OATP (SN-38) UGT1A9, -2B7 Changes in tubular surface area? Tubular secretion Transporter expression/activity Inhibitory effect of uremic solutes Proximal tubule cell number Renal metabolism UGT? GI 2 Gastric emptying time ph Expression of CYPs? 1 FDA Renal impairment Guidance 2 Rowland Yeo Expert Rev Clin Pharmacol 2011 3 Nolin Am J Kidney Dis 2003 4 Scotcher AAPS J 2016 5 Hsueh Mol Pharm 2016 6 Fujita Pharm Res 2014 7 Zhao J Clin Pharmacol 2012 8 Barnes Eur J Clin Pharmacol. 2014

Changes in plasma protein binding in CKD Parameter Albumin (g/l) M F Hematocrit (%) M F Heathy 44.9 41.8 43.0 38.0 GFR <30 ml/min/1.73m 2 37.6 35.0 39.7 33.2 Other factors that may affect protein binding: Conformational changes in albumin structure/binding sites Competition for binding sites by uremic solutes Limited data suggest elevated -acid glycoprotein Important to measure fu in RI population for highly bound drugs 1 Rowland Yeo Expert Rev Clin Pharmacol 2011

Systematic evaluation of the CKD effect on CYPs CYP2D6 CYP3A4/3A5 CYP2D6-mediated clearance decreased in parallel with the severity of CKD No apparent relationship between the severity of CKD and CYP3A4/5- clearance Effect on CYP1A2, CYP2C8, CYP2C9 and CYP2C19 Poster Tan et al. Yoshida Clin Pharmacol Ther 2016

PBPK modelling of RI - nonrenal CYPmediated clearance Retrospective analysis 1 repaglinide, telithromycin, slidenafil Healthy s.d. CYP abundance in RI extrapolated from clinical data Bosutinib PBPK 2 Step wise PBPK model development and verification RI Virtual population: Severe RI s.d. i. Reduced GFR, kidney weight and Q R ii. Reduced hepatic CYP3A4 expression iii. Reduced serum albumin and hematocrit 1 Zhao J Clin Pharmacol 2012 2 Ono DMD 2017

Effect of CKD on OATP Decrease in clearance in parallel with CKD severity Challenges: Lack of binding data in RI subjects Overlap between CYP2C8 and OATP drugs Poster Tan et al. - ITCW and ASCPT PT-020

PBPK modelling of RI renally eliminated drugs System parameters for mechanistic kidney models - healthy vs. RI?

Tubular surface area accounting for microvilli (ml/ min) (cm/ min) (cm 2 ) CL,,, P Nephron tubule considered as a cylinder Both PT cells and Caco-2 cells have extensive microvilli (apical membrane) - surface area. LoH, DT and CD cells - sparse/ negligible microvilli Area = 2 rh x number nephrons Kriz (1981).The Am. J. Physiol. 241(1):R3. Welling and Welling (1988) J. Electron. Micr. Tech. 9; 171-85

Tubular surface area - collecting duct requires special consideration! CCD Cortical Collecting Ducts formed by merging of app. 10 tubules (i.e. 900,000 nephrons/ kidney 90,000 CCD/ kidney) OMCD No merging in Outer Medulla Collecting Ducts (i.e. 90,000 OMCD/ kidney) Inner Medulla Collecting Ducts undergo successive dichotomous fusions (i.e. 90,000 IMCD/ kidney 360 Ducts of Bellini/ kidney) IMCD Scotcher, Eur J Pharm Sci 2016

Minimal tubular reabsorption model IVIVE Scaling P app to CL R,int ph gradient (6.5-7.4) Transporter inhibitor cocktail CL R,int,i = Papp TSA i Regional differences in TSA and tubular flow Scotcher, Eur J Pharm Sci 2016

Performance of the mechanistic tubular reabsorption model in vivo data from healthy gmfe (% predicted within 3-fold of observed) Proximal tubule only No correction for microvilli Reabsorption model All drugs (n = 45) 2.17 (76%) 5.35 (27%) 1.96 (87%) Low F reab (n = 17) Medium F reab (n=12) High F reab (n = 16) 1.59 (94%) 5.02 (35%) 1.97 (88%) 1.44 (92%) 8.52 (17%) 1.90 (92%) 4.11 (44%) 4.03 (25%) 2.01 (81%) Proximal tubule can be used as surrogate for low-med F reab (<75%) Consideration of correct tubular surface area of key relevance

In vitro-in vivo extrapolation of active renal secretion Increasing system complexity Transfected cells HEK, HeLa Immortalised cells LLC-PK1, ciptec, HK-2 Primary cultured renal tubule cells Kidney slices Scaling of kinetic parameters REF (V max /CL int ) ft IC 50 /Ki Tubular surface area (P app ) Proximal tubule cell number (V max /CL int ) 30 209 million PTC/ g kidney Kidney weight (CL int ) Dose Venous Blood PBPK model Kidney Liver Lung Brain Heart Muscle Adipose Skin Bone Rest of body Stomach Spleen Pancreas Small intestine Arterial Blood Dos Kidney-on-a-chip? Scotcher et al, AAPS J, part I 2016

Implementation of transporter expression data in PBPK models Relative expression factor = OAT3 expression in vivo / OAT3 expression in vitro Emerging proteomic data for renal transporters/ugts Missing data: Large cohort of individuals and special populations Regional and species differences Expression vs. functional activity Current REFs estimated using plasma or urinary excretion data 5.3 - HEK-OAT3 (pemetrexed) 2.3 - HEK-OCT2 (metformin) 3 - HEK-MATE1 (metformin) Scotcher AAPS J 2016 Part II; Prasad Drug Metab Dispos 2016; Knights Br J Clin Pharmacol 2016; Posada Drug Metab Dispos 2015; Burt EJPS 2016

Renal PBPK models special populations System Parameters Populations Young adults Paediatrics Elderly Renal impairment Kidney weight/ volume Renal blood flow GFR Inter-study variability Nephron number Highly variable No change after birth Regional tubule length/ diameter Variability within and between studies Proximal tubule only (1 study) Mainly proximal tubule Limited reports (qualitative) PTCPGK Rat data only Limited reports (qualitative) Limited reports (qualitative) Transporter abundance * 1 study human (pooled HKM) 1 study rat Mouse/ rat data Limited rat data Data available (quantitative, human) Limited or conflicting data Scotcher et al AAPS J 2016

Mechanistic digoxin kidney model: prediction of CL R in moderate to severe renal impairment Lee et al, CPT (2014) Existing PBPK model for digoxin incorporates transport by P-gp in liver and intestine (Neuhoff et al, J Pharm Sci, 2013) Consider role of P-gp and OATP4C1 (uptake) in kidney Availability of clinical data in healthy, elderly and different stages of RI

Development, verification and application of digoxin mechanistic kidney model Model development Glomerular filtration Calculated using Cockcroft-Gault eq. from serum creatinine f u,p from meta-analysis of reported data CL PD Same value for all tubular regions Transporter kinetics 1.IVIVE 1.P-gp in vitro K m and V max, REF 2.OATP4C1 not successful 2. OATP4C1 CL int,t estimated using clinical CL R data (19 clinical studies, 214 healthy subjects) Model verification Simulated vs. observed digoxin plasma concentration- and urinary excretion-time profiles Model application Elderly Renal impairment Scotcher et al, JPET 2017

Digoxin mechanistic kidney model verification Plasma concentration Urinary excretion rate

Mechanistic digoxin kidney model: prediction of CL R in renal impairment Scenarios tested in digoxin model: 1. Reduction in GFR alone 2. Modification of both GFR and active secretion a. OATP4C1 expression per million proximal tubule cells* b. P-gp expression per million proximal tubule cells* c. proximal tubule cellularity (PTCPGK) d. OATP4C1 expression or proximal tubule cellularity proportional to changes in GFR * Reflects also transporter activity due to inhibition by uremic solutes Scotcher et al, JPET 2017, ASCPT Quantitative Pharmacology, PII-122

Prediction of digoxin CL R in moderate to severe renal impairment reduction in GFR Assumption: NO changes in active secretion in renal impairment Over-estimation of CL R in RI Scotcher et al, JPET 2017

Mechanistic digoxin kidney model: prediction of CL R in severe renal impairment Additional mechanisms considered: i) transporter expression or ii) number of tubular cells Change in GFR only (severe renal impairment; GFR = 15 30 ml/ min) CL ratio CL CL renal impairment healthy subjects

Mechanistic digoxin kidney model: prediction of CL R in severe renal impairment Additional mechanisms considered: i) transporter expression or ii) number of tubular cells Change in GFR only (severe renal impairment; GFR = 15 30 ml/ min) CL ratio CL CL renal impairment healthy subjects

Mechanistic kidney model for digoxin: renal impairment OATP4C1 abundance and PTCPGK parameters changed proportionally to the change in GFR from the population representative ----- GFR only ----- GFR+PTCPGK ------ GFR+OATP4C1 Scotcher et al, JPET 2017

Take home message Assumption that secretion does not change in renal impairment over-estimated digoxin CL R Different mechanisms considered for active secretion in RI-PBPK model Comparable NET effect on the predicted systemic exposure and CL R Predicted dynamics inside proximal tubule cells different implications for nephrotoxicity or transporter-mediated DDIs Integrated bottom up-top down approaches important for stepwise RI-PBPK model development and verification Enhanced clinical trial design/adequate clinical data

Acknowledgements Chris Jones Daniel Scotcher Andres Olivares-Morales Hitesh Mistry Amin Rostami Leon Aarons Malcolm Rowland Shiew-Mei Huang, Ping Zhao, Lei Zhang Mingliang Tan Kenta Yoshida Maria Posada, Stephen Hall CAPKR consortium http://research.bmh.manchester.ac.uk/capkr/