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

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

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

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

Department of Pharmaceutics, University of Washington, Seattle, Washington (VK and JDU)

PBPK modeling of renal impairment what is missing?

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

Welcome to the webinar... We will begin shortly

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

Transporters DDI-2018

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

Supplemental Materials

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

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

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

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

Mass spectrometry-based quantification of transporter proteins and metabolizing enzymes: an update on advantages and challenges

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

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

Drug Interactions, from bench to bedside

Regulation of renal drug transport under physiological and pathological conditions

PHA 5128 CASE STUDY 5 (Digoxin, Cyclosporine, and Methotrexate) Spring 2007

In Vitro and In Vivo Interaction Studies Between Lesinurad, a Selective Urate Reabsorption Inhibitor, and Major Liver or Kidney Transporters

Reduction of metformin renal tubular secretion by cimetidine in man

Renal Clearance. Dr. Eman El Eter

MODULE PHARMACOKINETICS WRITTEN SUMMARY

Evaluation of Drug-Drug Interactions FDA Perspective

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

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

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

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

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

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

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

Renal Physiology. April, J. Mohan, PhD. Lecturer, Physiology Unit, Faculty of Medical Sciences, U.W.I., St Augustine.

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

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

Impact of Substrate-Dependent Inhibition on Renal Organic Cation. Transporters hoct2 and hmate1/2-k-mediated Drug Transport and

Cryo Characterization Report (CCR)

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

Bristol-Myers Squibb HCV DAAs: Review of Interactions Involving Transporters. Timothy Eley. 21 May 2014

PHA5128 Dose Optimization II Case Study I Spring 2013

Drug Interactions Between Direct-Acting anti-hcv Antivirals Sofosbuvir and Ledipasvir and HIV Antiretrovirals

The kidney. (Pseudo) Practical questions. The kidneys are all about keeping the body s homeostasis. for questions Ella

Supporting Information

Detail features... 1

Department of Pharmaceutics, University of Washington, P.O. Box , Seattle, WA 98195, USA

Use of in vitro cell assays and noninvasive imaging techniques to reduce animal experiments in drug development

Quantitative Evaluation of the Effect of P-Glycoprotein on Oral Drug Absorption

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

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

Calibrated Quantification. Christopher M. Shuford Laboratory Corporation of America Holdings

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

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

Renal tubular secretion in chronic kidney disease: description, determinants, and outcomes. Astrid M Suchy-Dicey

Development and Validation of an UPLC-MS/MS Method for Quantification of Mycotoxins in Tobacco and Smokeless Tobacco Products

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

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

Assessing Renal Function: What you Didn t Know You Didn t Know

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

Intra-renal Oxygenation. in Human Subjects

The effect of telaprevir on the pharmacokinetics of CYP3A and P-gp substrates

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

Challenges. Benefits. Control of viral load in plasma. Drug-drug interactions. Adverse effects/drug toxicities. Delay in HIV drug resistance

Renal Transporters- pathophysiology of drug - induced renal disorders. Lisa Harris, Pharmacist, John Hunter Hospital, Newcastle, 2015 November

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

Renal Physiology Part II. Bio 219 Napa Valley College Dr. Adam Ross

NORMAL POTASSIUM DISTRIBUTION AND BALANCE

Copyright 2009 Pearson Education, Inc. Copyright 2009 Pearson Education, Inc. Figure 19-1c. Efferent arteriole. Juxtaglomerular apparatus

HEK293 cells transfected with human MATE1, MATE2-K, or vector control were established by

Na + Transport 1 and 2 Linda Costanzo, Ph.D.

New issues in management of drug-drug interactions

BODY FLUID. Outline. Functions of body fluid Water distribution in the body Maintenance of body fluid. Regulation of fluid homeostasis

Clinical Pharmacology of DAA s for HCV: What s New & What s In Pipeline

Functions of Proximal Convoluted Tubules

Discovery and Validation of Pyridoxic Acid and Homovanillic Acid as Novel. Endogenous Plasma Biomarkers of Organic Anion Transporter (OAT) 1 and

Case Study 2 Answers Spring 2006

COMPETING INTEREST OF FINANCIAL VALUE

Kidney and urine formation

Water Reabsorption and the Effect of Diuretics on Urine Formation Patricia J. Clark, Ph.D. Department of Biology, IUPUI

RENAL FUNCTION An Overview

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

Mechanistic Modeling of Pitavastatin Disposition in Sandwich-Cultured Human Hepatocytes: A Proteomics-Informed Bottom-Up Approach s

Unit 2b: EXCRETION OF DRUGS. Ms.M.Gayathri Mpharm (PhD) Department of Pharmaceutics Krishna Teja Pharmacy college Subject code: 15R00603 (BPPK)

MEDICATIONS IN THE KIDNEY

Quantitation of Protein Phosphorylation Using Multiple Reaction Monitoring

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

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

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

Name: UFID: PHA Exam 2. Spring 2013

Overview of Intestinal Drug Metabolism and Transport

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

OST Course Schedule

General Principles of Pharmacology and Toxicology

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

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

Renal Pharmacology. Diuretics: Carbonic Anhydrase Inhibitors Thiazides Loop Diuretics Potassium-sparing Diuretics BIMM118

METFORMIN HYDROCHLORIDE - metformin hydrochloride tablet, extended release TEVA Pharmaceuticals USA Inc

1. remove: waste products: urea, creatinine, and uric acid foreign chemicals: drugs, water soluble vitamins, and food additives, etc.

DIAGNOSIS AND MANAGEMENT OF DIURETIC RESISTANCE. Jules B. Puschett, M.D.

Transcription:

Proteomic Quantification of Kidney Transporters: Methodological Challenges, Interindividual Variability and Application in IVIVE Bhagwat Prasad, Ph.D. University of Washington, Seattle, WA (bhagwat@uw.edu)

Drug transporters in proximal renal tubules About 30% of approved drugs are predominantly (>50% of total body clearance) cleared by the kidneys Brater, 2002; Feng et al., 2010 2

Renal Transporters and Clinical DDIs Transporters Inhibitor Victim ROA Change Reference OATs Probenecid Furosemide Iv AUC; Clr Smith et al., 1980; p.o AUC; Cl Vree et al., 1995 OCT2, MATEs Probenecid Cephradine Iv AUC; Clr Roberts et al., 1981 Cimetidine Metformin p.o. AUC; Clr Somogyi et al., 1987 Cimetidine Ranitidine p.o. AUC; Clr van Crugten et al., 1986 Cimetidine Dofetilide p.o. AUC; Clr Abel et al., 2000 Cimetidine Pindolol p.o. AUC; Clr Somogyi et al., 1992 Trimethoprim Procainamide p.o. AUC; Clr Kosoglou et al., 1988 P-gp Ritonavir Digoxin i.v. AUC; Clt, Clr Ding et al., 2004 Quinidine Digoxin i.v. Cserum ; Clt,Clr Leahey et al., 1981; Fenster et al., 1982, 1984 3

FDA Guidance on Renal Transporters Investigational drug Renal active secretion major? e.g., 25% of total Cl or unknown Determine whether investigational drug is an OAT1, OAT3 and/or OCT2 substrate in vitro PBPK modeling if an in vivo study is needed An investigational drug also should be evaluated to determine whether it inhibits OAT1, OAT3 and OCT2 MATE transporters should be considered when appropriate P-gp and BCRP are also investigated 4

Renal Transporters and Nephrotoxicity Need to predict tissue concentration 5

Knowledge gaps/challenges Good in vitro models are available However, conventional approaches to predict transporter mediated in vivo CL are not optimum lack of transporter protein abundance data interindividual variability in renal transporter activity is unknown 6

Challenges using alternate approaches 1. Poor correlation of mrna data with activity 2. Primary human proximal tubular cells (2D culture) tissue 7

Hypothesis Transporter quantification data can help predict transporter activity To translate in vitro transporter CL data to in vivo (IVIVE) To predict interindividual variability in renal transporter activity Transporter quantification approach Quantitative LC-MS/MS proteomics 8

Methodology: Quantitative Proteomics Surrogate peptide(s) generated from protein digestion is quantified LC-MS compatible sample LC-MS/MS Protein isolation Protein in solution Protein digestion (trypsin) Sample cleaning and enrichment MRM analysis of surrogate peptide Calibrator: Synthetic peptide standard Internal standard: Heavy labeled peptide standard

Is method development challenging? Not anymore QPrOmics TM (www.qpromics.uw.edu) *Poster (Prachi Jha) 10

Human kidney samples Non-cancerous portion of the human kidney cortex from nephrectomies UW medical center (n=20) Cortices of kidneys initially targeted for transplant purposes, but eventually not transplanted Ardea Biosciences (n=7) Newcastle University (n=14) 11

Renal transporters are homogenously expressed in kidney cortex 12

Transporter expression (pmol/mg membrane protein) Renal transporter expression and inter-individual variability 100 10 1 0.1 OAT1 OAT3 OAT2 OAT4 OCT2 OCTN1 OCTN2 P-gp MRP2 MRP4 MATE1 SGLT2 13

Renal Transporter Pie 3% 2% 2% OCT2 OAT1 4% 3% MATE1 26% 5% OAT3 P-gp 7% MRP2 OCTN1 12% 18% OAT2 MRP4 OCTN2 18% OAT4 14

Pmol/mg total membrane protein Correlation of renal transporters 15

Pmol/mg total membrane protein SGLT2 expression correlates with majority of renal drug transporters 16

Application of transporter expression data to predict metformin secretory CL Excretion Urine (>90%) CL(Renal) CL (secretory) 552 ml/min 432 ml/min 17

OCT2 expression in human kidney cortex and in OCT2 expressing HEK and MDCKII cells Data generated by Vineet Kumar (Unadkat Lab) 18

Transporter expression based scaling factor CL = V max K m and V max = K cat * [T] Scaling Factor = [T] in vivo [T] in vitro OCT2 Expressing Cells OCT2 Expression (fmol/µg protein) [ 14 C] Metformin Uptake Activity (pmol/min/mg protein) HEK293 369.4 ± 26.8 202.1 MDCKII 19.0 ± 1.1 10.9 HEK293/MDCKII 19.4 18.5 Data generated by Vineet Kumar (Unadkat Lab) in collaboration with Dr. Joanne Wang and Jia Yin 19

IVIVE of metformin CL Sec Human kidney weight (g) 150 Cortex WT/kidney (70% of total kidney weight) (g) 105 OCT2 expression in cells (fmol/µg Protein) 369.4 (HEK293) 19 (MDCKII) OCT2 expression in Kidney (fmol/µg Protein) 7.6 In-vitro Clearance (µl/mg protein/min) 36.7 (HEK293) 2.0 (MDCKII) Scaling factor (SF) 0.02 (HEK293) 0.40 (MDCKII) mg of protein per unit cortex weight 0.3 CL Sec = 2 x CLin vitro x E in vivo x protein per unit cortex weight x cortex weight E in vitro Metformin CL Sec (ml/min) 47.6 (HEK293) 50.4 (MDCKII) Renal secretory clearance of metformin: 432 ml/min Kumar et al. (manuscript) 20

% Expressed on plasma membrane A substantial % of OCT2 protein is localized in the intracellular (inactive) fractions 90 80 70 60 50 40 30 20 10 0 HEK Cells MDCKII Cells OCT2 Na K ATPase Calreticulin HEK293 MDCKII Metformin CL Sec, ml/min (uncorrected) 47.6 50.4 Metformin CL Sec, ml/min (Corrected for intracellular contamination) 148.2 97.7 In vivo observed renal secretory clearance of metformin: 432 ml/min Kumar et al. (manuscript) 21

Other possible reasons of underprediction 1. Mechanisms of OC transport Proximal tubular cell Blood Plasma membrane potential in cell lines OC + -70 mv OCT2 OC + HEK293 cells: -19 to -27 mv MDCKII cells: -20 to -50 mv -Chien et al., DMD, 2016 2. Role of other (unknown?) transporters Kumar et al. (manuscript) 22

Conclusions OCT2, OAT1, OAT2 and MATE1 are the major drug transporters in human kidneys A significant protein-protein correlation was observed between renal transporters The transporter protein expression and correlation will be useful for the PBPK prediction of renal drug disposition including predicting tissue concentration A significant fraction of transporters can be present in the intracellular pools in the over-expressing cell lines The predicted metformin CL Sec is 30% of the observed in vivo Cl sec using the transporter expression based scaling (when correlated for intracellular contamination) Importance of mechanism of drug transport in IVIVE 23

Acknowledgements University of Washington, Seattle, WA Jashvant Unadkat, Ph.D. Jonathan Himmelfarb, M.D. Ed Kelly, Ph.D. Joanne Wang, Ph.D. Other significant contributors Vineet Kumar Katherine Johnson Sarah Billington, Ph.D. Jia Yin Gabby Patilea-Vrana Newcastle University, UK Colin Brown, Ph.D Ardea Bio Caroline Lee, Ph.D Grants UH2TR000504 DA032507 Ardea Bio UWRAPT 24

UWRAPT Drs. Cornelis Hop (Marcel) & Laurent Salphati Genentech Drs. Raymond Evers & Xiaoyan Chu Merck Drs. Guangqing Xiao & Chuang Lu Biogen Drs. Yurong Lai and Wenying Li BMS Drs. Donavon McConn and Mingxiang Liao Takeda Drs. Brian Kirby, Adrian Ray and Anita Mathias Gilead Drs. Caroline Lee & Ravindra Alluri Ardea Bio & AZ Dr. Anshul Gupta, AZ 25