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

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

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

PHA5128 Dose Optimization II Case Study I Spring 2013

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

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

104 MMWR December 17, 2004

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

Evaluation of Drug-Drug Interactions FDA Perspective

Supplemental Materials

Monica Edholm Medica Medic l a Pr oducts Agency

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

Welcome to the webinar... We will begin shortly

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

Transporters DDI-2018

PHA First Exam Fall 2003

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

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

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

General Principles of Pharmacology and Toxicology

Renal Impairment From Dettli to Guideline: What can we learn?

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

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

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

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

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

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

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

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

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

Current Drugs: Drug-Drug Interactions

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

Renal Clearance. Dr. Eman El Eter

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

General Principles of Pharmacology and Toxicology

Drug Interactions, from bench to bedside

PBPK modeling of renal impairment what is missing?

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

RISK FACTORS AND DRUG TO STATIN-INDUCED MYOPATHY

New issues in management of drug-drug interactions

Effects of Renal Disease on Pharmacokinetics

MODULE PHARMACOKINETICS WRITTEN SUMMARY

Patient details GP details Specialist details Name GP Name Dr Specialist Name Dr R. Horton

ASSESSMENT OF THE ROLE OF SOLUTE CARRIER DRUG TRANSPORTERS IN THE SYSTEMIC DISPOSITION OF FLUOROQUINOLONES: AN IN VITRO - IN VIVO COMPARISON

BASIC PHARMACOKINETICS

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

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

PHA First Exam. Fall 2004

Endogenous Biomarkers for OATP1B: Preclinical to Clinical Translations. Yurong Lai, PhD Drug Metabolism Gilead Sciences

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

Reduction of metformin renal tubular secretion by cimetidine in man

Abemaciclib Inhibits Renal Tubular Secretion Without Changing Glomerular Filtration Rate

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

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

1. If the MTC is 100 ng/ml and the MEC is 0.12 ng/ml, which of the following dosing regimen(s) are in the therapeutic window?

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

Click to edit Master title style

toxins ISSN

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

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

PRESCRIBING IN LIVER AND RENAL DISEASE

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

Multiple Interactions of Cimetidine and Probenecid with Valaciclovir and Its Metabolite Acyclovir

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

12/9/2015. Drug Interactions. Sarah Robertson, Pharm.D. Director, Department of Clinical Pharmacology Vertex Pharmaceuticals Inc.

Renal Function and Associated Laboratory Tests

Renal Excretion of Drugs

Section 5.2: Pharmacokinetic properties

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

Levocetirizine dihydrochloride

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

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

Antiretroviral Dosing in Renal Impairment

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

TDF Renal Dysfunction

Pharmacokinetics for Physicians. Assoc Prof. Noel E. Cranswick Clinical Pharmacologist Royal Children s Hospital Melbourne

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

The principal functions of the kidneys

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

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

HIV AND ANTIRETROVIRALS: A DISPENSERS PERSPECTIVE WITH A FOCUS ON DRUG INTERACTIONS DR. M.M. PEZA MBCHB (UCT) PG DIPLOMA (HEALTH ECONOMICS) (UCT)

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

Appendix 4: Renal impairment

saxagliptin / metformin HCl

Integrase Strand Transfer Inhibitors on the Horizon

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

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

STRIBILD (aka. The Quad Pill)

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

Renal-Related Questions

Filtration and Reabsorption Amount Filter/d

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

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

Exploiting BDDCS and the Role of Transporters

Physiology Lecture 2. What controls GFR?

PHAR 7632 Chapter 16

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

BIOL 2402 Renal Function

Module Summary of Product Characteristics

MODELING MECHANISM BASED INACTIVATION USING PBPK JAN WAHLSTROM DIRECTOR, PRECLINICAL

Transcription:

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

Outline Background of renal transporters. Clinically observed transporter-mediated renal DDIs. Quantitative prediction of renal DDIs. A case study to predict renal DDIs. Renal transporter inhibition and elevated serum creatinine.

Renal Clearance If CLr > fu*gfr, transportermediated active secretion must be involved. Transporter interactions could lead to clinical DDIs. CLr = fu*gfr+ CL secretion - CL reabsorption

Major Human Renal Transporters Blood Flow net secretion CL r > fu*gfr OAT1 OAT3 OCT2 Amino acid OAT4 MRP2/4 MDR1 OCTN1 H+ OCTN2 MATE1/2-K PepT2 L-Carnitine Filtration (GFR) net reabsorption CL r < fu*gfr Proximal Tubular Cell Urine CL r = fu*gfr+ CL secretion - CL reabsorption

Outline Background of renal transporters. Clinically observed transporter-mediated renal DDIs. Quantitative prediction of renal DDIs. A case study to predict renal DDIs. Renal transporter inhibition and elevated serum creatinine.

Renal Transporter-mediated DDIs Given the limitations of dosage and inhibitor concentrations relative to the inhibition potency against the transporters Cimetidine is one of the most potent OCT2 inhibitors in vivo. Probenecid is claimed as the most potent OATs inhibitor in vivo.

Renal Transporter-mediated DDIs The magnitude of renal clearance reduction caused by cimetidine is lower compared with the renal clearance reduction from probenecid The higher inhibition potency of probenecid compared with cimetidine. The magnitude of AUC changes from renal DDIs is much lower than the AUC changes from CYP DDIs and hepatic OATPsmediated DDIs Despite the fact that most of the market drugs have significant renal clearance, even complete inhibition of transporter is assumed, most renal DDIs are predicted to be <2-fold Renal impairment causes weak or moderate effects.

Why AUC Changes from Renal DDIs are Small? About 30% of drugs have renal clearance as a major elimination pathway. Why don t we observe significant renal DDIs due to the inhibition of renal transport? No potent renal transporter inhibitors exist? Renal clearance mechanism protect from DDIs, fraction of clearance inhibitable is low? Significant renal DDIs have not been systemically documented?

Outline Background of renal transporters. Clinically observed transporter-mediated renal DDIs. Quantitative prediction of renal DDIs. A case study to predict renal DDIs. Renal transporter inhibition and elevated serum creatinine.

Static Models to Predict DDIs Basic model Mechanistic model

Renal OATs DDIs by Probenecid Compare probenecid inhibition potency towards hoat1 and hoat3 (Ki ~8 mm and ~3 mm, respectively) with unbound plasma concentration of probenecid. Using CLsec,i = CLsec/(1+I/Ki), it is reasonable to estimate that probenecid at 2000 mg dose (unbound C max ~3-50 mm) is able to inhibit about 80% of renal transport of victim drugs through OATs in vivo. Example: Victim drug A and probenecid DDI Drug A: Control CLr, CLsec = CLr - fu*gfr With probenecid, CLr,i = fu*gfr + CLsec x 0.2 Compare with the observed renal clearance of drug A with probenecid in clinical DDI trial

IVIVC of OATs-mediated Renal DDIs Victim Perpetrator Control renal clearance Renal clearance with (ml/min) perpetrator (ml/min) Renal clearance reduction Predicted renal clearance reduction Acyclovir Probenecid 248 168 32% 47% Bumetanide Probenecid 145 22.0 85% 79% Cefamandole Probenecid 229 57.0 75% 60% Cefmenoxime Probenecid 159 66.0 58% 44% Cidofovir Probenecid 151 95.7 36% 21% Cimetidine Probenecid 360 270 25% 58% Cinoxacin Probenecid 153 66.0 57% 61% Ciprofloxacin Probenecid 373 134 64% 65% Enalapril Probenecid 229 61.0 73% 61% Enalaprilat Probenecid 108 66.0 39% 25% Famotidine Probenecid 297 107 64% 73% Fexofenadine Probenecid 230 74.0 68% 65% Furosemide Probenecid 72.8 20.3 72% 78% Ganciclovir Probenecid 235 190 19% 40% Nafcilin Probenecid 141 39.2 72% 73% Oseltamivir Probenecid 262 95.0 64% 44% Zalcitabine Probenecid 310 180 42% 50% Zidovudine Probenecid 333 209 37% 58% Feng, Mol. Pharmaceutics 2013

Renal OCT DDIs through Cimetidine Compare cimetidine inhibition potency towards OCT2 (Ki ~ 30 mm) with unbound plasma concentration of cimetidine (unbound C max ~8.6-73 mm). Using CLsec,i = CLsec/(1+I/Ki), it is reasonable to estimate that cimetidine is able to inhibit about 30% or more of renal transport of victims through OCT2 in vivo.

IVIVC of OCT2-mediated Renal DDIs Victim Perpetrator Control renal clearance (ml/min) Renal clearance with perpetrator (ml/min) Renal clearance reduction Predicted renal clearance reduction Acyclovir Cimetidine 349 273 22% 22% Amiloride Cimetidine 358 299 16% 24% Cephalexin Cimetidine 263 208 21% 18% Dofetilide Cimetidine 274 238 or 184 13%-33% 25% Fexofenadine Cimetidine 230 152 34% 25% Metformin Cimetidine 728 403 45% 25% 527 378 28% 23% Procainamide Cimetidine 466 297 36% 24% Cimetidine 202 130 36% 15% Cimetidine 347 196 43% 21% Ranitidine Cimetidine 326 244 25% 21% Varenicline Cimetidine 133 100 25% 8.0% Zidovudine Cimetidine 478 210 56% 25% Feng, Mol. Pharmaceutics 2013

Basic Model to Predict renal Clearance Reduction The basic model reasonably predicted the changes in CLr of OAT1/OAT3 and OCT2 substrates, when concomitantly dosed with probenecid and cimetidine, respectively. The prediction renal clearance changes with probenecid for 11 of 18 (61%) cases are within 25% error, and 17 of 18 (94%) cases are within 50% error of the observed CLr changes. The predicted renal clearance changes with cimetidine for 8 of 13 (61%) cases were within 25% and 12 of 13 (92%) cases within 50% of the observed CLr changes. The basic model accounts for the inhibitory effect of a perpetrator at the high end of clinical relevant exposure range to predict the change in renal clearance of the victim drug. Feng, Mol. Pharmaceutics 2013

Mechanistic Model A comprehensive mechanistic model was developed to examine the effect of inhibition of renal secretion transporters on plasma exposures of victim drug. This is principally similar to Rowland Matin equation proposed for prediction of CYP-related DDIs. The mechanistic model takes into consideration the importance of renal clearance relative to the total clearance of a victim and the variation of a perpetrator s concentration to predict the change in AUC of the victim.

Mechanistic Model Where CL and CLi are total systemic clearance in the absence and presence of inhibitory drug, respectively. CLsec,c represents secretory clearance in control. CLx represents nonsecretory clearance (CLx = CL CLsec,c). Feng, Mol. Pharmaceutics 2013

Mechanistic Model to Predict AUC Changes The mechanistic model predicted 7 of 13 (54%) cases within 25% and 12 of 13 (92%) cases within 50% of the observed AUC ratios with probenecid. Feng, Mol. Pharmaceutics 2013

AUC Changes as a Function of CLsec/CL at various [I]/Ki AUC change from renal DDI is dependent on renal secretion pathway and inhibitor potency. Feng, Mol. Pharmaceutics 2013

Summary from In vitro In vivo Analysis of Renal DDI Cimetidine is not only a renal OCT2 inhibitor, but also a potent inhibitor of MATEs. Prediction of cimetidine DDIs need to consider both OCT2 and MATEs inhibitions. The extent of renal clearance change of the affected victim drug can be predicted from the in vitro IC 50 of the inhibiting perpetrator drug and its unbound therapeutic concentrations in the plasma. Based on the positive relationship from in vitro in vivo renal DDI analysis, it is confident to predict in vivo renal DDIs based on in vivo PK and in vitro transporter interaction.

Conclusions If a NCE is identified as a substrate of renal transporters, its renal clearance reduction and AUC change with transporter inhibitors can be predicted with its human renal clearance and other PK parameters. If re-absorption process is involved in the renal clearance Underestimate the contribution of active secretion in renal clearance Underestimate renal clearance reduction in vivo. Many drugs may be handled by multiple transporters with overlapping substrate and inhibitor specificities. The reduction of renal active secretion due to the inhibition of a transporter may be overestimated. The magnitude of AUC change caused by renal DDIs is dependent on the contribution of active secretion clearance in total clearance, as well as inhibitor potency compared with systemic concentration. Clinical significant interactions occur only when the affected transporter represents the major pathway for overall elimination.

Conclusions (con t) Because of the involvement of multiple renal processes including filtration, tubular secretion and tubular reabsorption in renal drug handling, and the functional redundancy of some renal drug transporters, sever clinical DDIs at the renal level seem to be not very common. However, clinical relevance of renal DDIs needs to be evaluated in the context of efficacy and safety profile of the affected drug. Awareness of the possibility of transporter-mediated DDIs is necessary for drugs that are on the current market and for compounds in clinical development. To better predict transporter-mediated DDIs from in vitro data, the quantitative contribution of each individual transporter to the total drug clearance in vivo needs to be determined.

Outline Background of renal transporters. Clinically observed transporter-mediated renal DDIs. Quantitative prediction of renal DDIs. A case study to predict renal DDIs. Renal transporter inhibition and elevated serum creatinine.

Case Study : Prediction of renal DDIs associated with OAT1 inhibition by Compound A Unchanged renal excretion is the major route of elimination for compound A in humans. >90% of the dose was excreted unchanged in urine in clinical studies Transporter Inhibition. No inhibition was observed up to 1000 μm for P-gp/BCRP/OAT3/OCT2/OATP1B1/OATP1B3 OAT1 Inhibition IC 50 = 281 μm Static DDI Estimation. Clinical Plasma Concentrations Loading Dose free C max = 43 µm Maintenance Dose C max = 26 µm Free C max /IC 50 0.15 (>0.1) based on loading dose 0.09 based on maintenance dose

Change in renal CL of victim drug AUC ratio of victim drug Predicted change in Renal clearance and plasma AUC of OAT1 substrate drugs when co-dosed with Probenecid 1.00 4 0.80 3 0.60 0.40 2 0.20 1 *ND *ND *ND *ND *ND *ND Not determined due to lack of appropriate substrate information Probenecid inhibit OAT1/3 and show maximum reported renal DDIs. A maximum AUCR of ~3 was predicted for naficillin as victim drug, and cimetidine, furosemide and zidovudine show ~2, while majority of the interactions are predicted to be <2-fold. Mechanistic models were validated with the clinical data.

Change in renal CL of victim drug AUC ratio of victim drug Predicted change in Renal clearance and plasma AUC of OAT1 substrate drugs when co-dosed with compound A 1.00 1.25 0.95 1.20 0.90 1.15 0.85 1.10 0.80 1.05 0.75 1.00 *ND *ND *ND *ND *ND *ND Not determined due to lack of appropriate substrate information In relation to DDIs noted with probe inhibitors (probenecid), compound A may show no impact on both the changes in plasma AUC and Renal CL of OAT1 substrates. A maximum AUCR of ~1.25 was predicted for naficillin as victim drug, while majority of the interactions are predicted to be <10% change. The risk of OAT1-mediated interactions by compound A was predicted to be minimal.

Prediction of Renal DDI by Compound A Mechanistic renal DDI model predicts maximum interaction of ~25% AUC increase when co-administered compound A, while the majority of interactions with OAT1 substrates < 10% change. OAT1 substrates, e.g., zidovudine, acyclovir, tenofovir, methotrexate, etc. are not common co-medications for the patient population. OAT1 substrates are not narrow TI compounds, so no limitations to concomitant medication. Magnitude of know OAT1 DDIs is small. Probenecid OAT1 inhibitor- 3.6 x increase w/cephradine, 1.5 x increase w/cidofovir, 1.4 x increase w/acyclovir, 2.9x increase in Furosemide (both OAT3 and OAT1 ) No Clinical OAT1 DDI studies are planned.

Outline Background of renal transporters. Clinically observed transporter-mediated renal DDIs. Quantitative prediction of renal DDIs. A case study to predict renal DDIs. Renal transporter inhibition and elevated serum creatinine.

Creatinine Interacts with Renal Transporters Mathialagan, J Pharm Sci 2017

Renal Transporter Inhibition and Free Cmax C max,u /IC 50 Inhibitors C max,u (µm) hoct2 hmate1 hoat2 hmate2k Trimethoprim 3.9 0.151 2.407 0.039 6.724 Pyrimethamine 0.299 0.322 1.759 0.003 0.854 Cimetidine 9.60 0.264 2.540 0.094 0.405 Cobicistat 0.05 0.005 0.012 0.002 0.002 Vandetanib 0.03 0.055 0.115 0.000 0.158 Dolutegravir 0.13 0.016 0.022 0.001 0.003 Ranolazine 2.28 0.307 0.410 0.021 0.041 Dronedarone 0.006 0.003 0.003 0.000 0.001 Amiodarone 0.09 0.011 0.016 0.000 0.000 Ritonavir 0.03 0.003 0.071 0.004 0.001 Telaprevir 1.39 0.023 0.053 0.007 0.012 Famotidine 40mgs 0.312 0.011 1.156 0.002 0.043 Ranitidine 3.16 1.311 0.564 0.011 0.929 Nizatidine 8.2 0.522 1.763 0.115 0.082 Quinidine 0.52 0.033 0.026 0.002 0.066 Using free Cmax will generate false negatives and false positives Mathialagan, J Pharm Sci 2017

Renal Transporter Inhibition and Total Cmax Total C max /IC 50 Inhibitors Total C max (µm) hoct2 hmate1 hoat2 hmate2k Trimethoprim 6.9 0.267 4.259 0.069 11.897 Pyrimethamine 2.3 2.473 13.529 0.023 6.571 Cimetidine 12 0.331 3.175 0.117 0.506 Cobicistat 1.55 0.145 0.378 0.064 0.069 Vandetanib 0.33 0.600 1.269 0.004 1.737 Dolutegravir 13.1 1.588 2.259 0.131 0.266 Ranolazine 6.01 0.810 1.081 0.055 0.108 Dronedarone 0.3 0.145 0.163 0.001 0.058 Amiodarone 2.3 0.293 0.411 0.012 0.012 Ritonavir 2.16 0.214 5.143 0.298 0.052 Telaprevir 5.82 0.096 0.222 0.029 0.051 Famotidine 40mgs 0.39 0.014 1.444 0.002 0.053 Ranitidine 3.72 1.544 0.664 0.012 1.094 Nizatidine 12.7 0.809 2.731 0.178 0.127 Quinidine 4 0.256 0.202 0.018 0.506 Using total Cmax will generate false positives Mathialagan, J Pharm Sci 2017

Summary Creatinine was used as the common substrate to assess inhibition potency against renal OCT2, MATE1, MATE2K and OAT2 transporters. When Cmax/IC50 0.1 is used as a cutoff, all of the 11 compounds with elevated serum creatinine can be attributed to the inhibition of at least one of the 4 renal transporters Unfortunately, the 4 negative controls without elevated serum creatinine also demonstrated inhibition of at least one renal transporter. Renal transporter inhibition studies are useful to rationalize the cases where elevated serum creatinine are observed in human. Assessment of renal function marker other than creatinine is helpful. Although creatinine is a substrate of OAT2 in vitro, more in vivo data are needed to gain a better understanding of the role of OAT2 in creatinine renal secretion in vivo.

Acknowledgments Manthena Varma Yasong Lu Sumathy Mathialagan David Rodrigues