Deuteration of Drugs for Pharmacokintic Enhancement: Considerations Essential for Success

Similar documents
Chapter 4. Drug Biotransformation

Deuterium Isotope Effects on Drug Pharmacokinetics I: System-Dependent Effects of Specific Deuteration with Aldehyde Oxidase Cleared Drugs.

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

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

B. Incorrect! Compounds are made more polar, to increase their excretion.

MODULE No.26: Drug Metabolism

INTRODUCTION TO PHARMACOKINETICS

NON-P450-MEDIATED METABOLISM: IDENTIFICATION AND IMPLICATION DURING COMPOUND SELECTION AND OPTIMIZATION

MEDCHEM 570. First Midterm. January 30, 2015

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

Section 5.2: Pharmacokinetic properties

Metabolism. Objectives. Metabolism. 26 July Chapter 28 1

Click to edit Master title style

Toxicant Disposition and Metabolism. Jan Chambers Center for Environmental Health Sciences College of Veterinary Medicine

METABOLISM. Ali Alhoshani, B.Pharm, Ph.D. Office: 2B 84

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

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

The Analysis of Isotopically Labeled Propylene Glycol in ecigarettes

Industrial Toxicology

BIOPHARMACEUTICS and CLINICAL PHARMACY

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

Public Assessment Report. Scientific discussion. Ropinirol Actavis. Ropinirole hydrochloride DK/H/1212/ /DC

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

XTreme 200 Human Liver Microsomes Lot No Human Liver Microsomes Pool of 200 (100 Male and 100 Female) Suspension medium: 250 mm sucrose

Orientation for New Child and Adolescent Psychiatry Residents: Module Four Pediatric Psychopharmacology

Biologic Oxidation BIOMEDICAL IMPORTAN

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

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

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

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

Aldehyde Oxidase and Donor Variability Challenges to Estimating Human Clearance

III. TOXICOKINETICS. Studies relevant to the toxicokinetics of inorganic chloramines are severely

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

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?

Human ADME Study Design Considerations in Healthy Subjects and in Patients

The Opportunity: c-ibs and pain relief with confidence YKP10811

Exploiting BDDCS and the Role of Transporters

Metabolic Changes of Drugs and Related Organic Compounds

Chapter 9. Biotransformation

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

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

2. List routes of exposure in the order of most rapid response.

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

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

Involvement of CYP2C8 and UGT1A9 in the metabolism of a novel gastroprokinetic agent, Z-338

Influence of fluvoxamine on carvedilol metabolism and plasma disposition in vitro and in vivo experiments

Metabolic Changes of Drugs and Related Organic Compounds

A novel microdose approach to assess bioavailability, intestinal absorption, gut metabolism, and hepatic clearance of simeprevir in healthy volunteers

Drug Metabolism Phase 2 conjugation reactions. Medicinal chemistry 3 rd stage

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

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

Principles of Toxicokinetics/Toxicodynanics

Phase I in clinical drug development

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

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

Mechanism of Detoxification

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

Absolute bioavailability and pharmacokinetic studies in early clinical development using microdose and microtracer approaches.

Six Types of Enzyme Catalysts

Waters ASMS Users Meeting May 30 th, Overview

Introduction to. Pharmacokinetics. University of Hawai i Hilo Pre-Nursing Program NURS 203 General Pharmacology Danita Narciso Pharm D

Take-Home Exam Distributed: October 16, 2013, at 1:30 p.m. Due: October 21, 2013, at 10:00 a.m.

Lippincott Questions Pharmacology

AMS: A smarter way to cut through the MIST. Bob Ings RMI-Pharmacokinetics

1. Measurement of the rate constants for simple enzymatic reaction obeying Michaelis- Menten kinetics gave the following results: =3x10-5 = 30μM

Dr. Nafeth Abu-Tarbou sh Introduction to Biochemist ry 15/08/2014 Sec 1,2, 3 Sheet #21 P a g e 1 Written by Baha Aldeen Alshraideh

Edgar Naegele. Abstract

Nonlinear Pharmacokinetics

Sylvie Klieber, Catherine Arabeyre-Fabre, Patricia Moliner, Eric Marti, Martine Mandray, Robert Ngo, Celine Ollier, Priscilla Brun & Gerard Fabre

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

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

Aug 28 th, 2017 Pierre Daublain

Chapter 11: Enzyme Catalysis

3) How many different amino acids are proteogenic in eukaryotic cells? A) 12 B) 20 C) 25 D) 30 E) None of the above

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

BASIC PHARMACOKINETICS

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

CONTENT. i iv ix. SVKM s NMIMS, School of Pharmacy and Technology Management

Objectives Making CYP450, Drug Interactions, & Pharmacogenetics Easy

Porphyrins: Chemistry and Biology

10th French-Belgian ABC Meeting Brussels, October, 2012

1/3/2011. Chapter 17 Carboxylic Acids and Their Derivatives. Nucleophilic Addition- Elimination at the Acyl Carbon

MS/MS as an LC Detector for the Screening of Drugs and Their Metabolites in Race Horse Urine

Pharmacokinetics, pharmacodynamics and pharmacogenetics of aripiprazole and olanzapine in healthy subjects

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

Margaret A. Daugherty. Announcements! Fall Michaelis Menton Kinetics and Inhibition. Lecture 14: Enzymes & Kinetics III

BACKGROUND AND PURPOSE

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

number Done by Corrected by Doctor

Mass Spectrometry Introduction

Mohammad Alfarra. Faisal Al Nemri. Hala Al Suqi

MODELING MECHANISM BASED INACTIVATION USING PBPK JAN WAHLSTROM DIRECTOR, PRECLINICAL

CYP2D6: mirtazapine 2001/2002/2003

Pharmacokinetics. Karim Rafaat

Rational Dose Prediction. Pharmacology. φαρμακον. What does this mean? pharmakon. Medicine Poison Magic Spell

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

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

Pharmacokinetics of Rifampicin in African Children Evaluation of the new WHO dosing guidelines

Chapter 18 Carboxylic Acids and Their Derivatives. Nucleophilic Addition- Elimination at the Acyl Carbon

f FAD C-4a-hydroperoxy form of FMO enzymes 131 flavin monooxygenases 241

Transcription:

Deuteration of Drugs for Pharmacokintic Enhancement: Considerations Essential for Success Alvin D.. Vaz Alfin D. Vaz Pfizer Global Research and Development Groton, CT, USA Collaborators: Raman Sharma, Tim Strelevitz, Aarti Sawant, Alan Clark, Elaine Tseng, Hongying Gao, Klass Schildnegt, Patrick Verhoest, Vinod Parikh,

Uses for Deuterated Drugs As an internal standard in quantitative analysis Three or more non-exchangeable deuterium atoms incorporated to mass-differentiate the internal standard and analyte Alter pharmacokinetic or toxic properties Deuterium substitution for hydrogen does not appreciably change physicochemical properties such as polar surface area, molecular volume, hydrogen bonding, is naturally abundant to 0.09%, 09% and is OT radioactive. One or more deuterium atoms substituted t at specific sites can slow metabolic clearance and result in: Increases in C max, drug exposure (AUC), and systemic half- life (T 1/2 ) Decreased metabolically generated toxic metabolites

Origin of the Kinetic Deuterium Isotope Effect (KDIE) Difference in mass between deuterium and hydrogen results in a zero-point energy difference between C-H and C-D bonds, consequently an increase in energy required to break a C-D bond If the transition state involves a symmetrical breaking of a C-H bond, substitution of hydrogen by deuterium slows down the reaction rate by a factor of 5 9 KDIE = k H /k D ~ 5 _ 9 Enzyme intrinsic isotope effect = D k 1 / H k 1 The intrinsic isotope effect reflects the enzyme s commitment to catalysis In pharmacokinetics the intrinsic clearance isotope effect reflects the In pharmacokinetics the intrinsic clearance isotope effect reflects the kinetic isotope effect on the first order rate constant for disappearance of substrate or V max /K m

Press releases by Concert Pharmaceuticals 43% increase in preclinical (monkey) half life (4.5 to 6.3 hrs) for deuterated Linezolid Oct 27th 2008 Phase 1 start for deuterated Paroxetine Sept. 25th, 2008; announced protection against CYP2D6 inactivation Sept. 29th, 2009 Phase 1b multiple dose escalation study for deuterated Atazanavir ov 9th, 2009 Expectation: QD dosing without Ritonavir co-administration Effect of deuterium substitution on sympathomimetic amines on adrenergic responses. Belleau, B.; Burba, J.; Pindell, M.; Reiffenstein, J. Science (1961), 133 102-4.

Systemic Clearance Mechanism and KDIEs on Pharmacokinetics CL systemic = CL H + CL extrahepatic CL urine + CL other Clearance enzymes where KDIEs apply Aldehyde Oxidase 4 6 Monoamine Oxidases 2 9 Cytochromes P450 1 9 Alcohol/aldehyde CL metabolic + CL bile Clearance enzymes where KDIEs do not apply Flavin monooxygenases Glucuronyl transferases Sulfotransferases Glutathione-S-transferases dehydrogenases 6 8 -acetyl trasferases For pharmacokinetic enhancement: Metabolic clearance must determine systemic clearance The enzyme(s) involved must have KDIEs on their intrinisic clearance

Why KDIEs with aldehyde oxidase? Aldehyde Oxidase (AO) is a cytosolic molybdopterin class oxidase, hydroxylates nitrogen heterocycles to the nitrogen Experience with human PK failure due to high clearance by AO Interspecies variability that results in failure of allometric scaling Lack of direct in vitro to in vivo correlation in clearance scaling possibly due to wide tissue distribution Proposed reaction mechanism involves a rate-limiting hydride/proton abstraction Potential to alter PK by use of KDIE

KDIEs to establish interspecies commonality in the AO reaction mechanism Intra-molecular isotope effect (intrinsic) H H Spectra of hydroxylated y metabolite of pthalazine after 30 minute incubation in guinea k H pig/ human cytosol D H k D 148.10 100 OH D 90 H 1-2 H-Phthalazine 80 5:1 ratio D/H H O 70 H m/z 148 D H 60 O Intensity 50 40 30 20 10 0 m/z 147 OH H 147.10 149.10 140.15 141.16 150.14 152.18 157.18 159.10 161.14 140 142 144 146 148 150 152 154 156 158 160 m/z H D k H k D 2-2 H-Quinoxaline O H M+H - 148 amu k H /k D = m/z148 / m/z147 M+H = 147 amu M+H - 148 amu D k H /k D = m/z148 / m/z147 H O M+H = 147 amu Substrate H k / D k with cytosolic AO from Guinea pig Rat human 2-2 H-Quinoxaline 4.7 5.1 5.0 2-2 H-Phthalazine 4.9 5.0 5.1

Inter-molecular KDIE on competitive first order elimination rate constants Substrate H k / D k with cytosolic AO from human rat guinea pig Quinoline 5.5 6.1 6.0 Carbazeran 4.8 5.0 6.0 Zoniporide 5.8 3.6 4.8

Inter-molecular KDIE on steady state kinetic constants v/s 250 200 inolone l/min/ml 2-qu pmol 150 100 50 0 Vmax 5:1 0 200 400 600 800 1000 1200 Quinoline [um] Quinoline 2-2 H-Quinoline K m (mm) 212 193 V max (pmol/min) 246 47 V max / K m 1.2 0.2 KDIE on Cl int = 6.0

Conclusions from in-vitro KDIEs for AO- catalyzed reactions Across species the rate-limiting step in AO-catalyzed reactions is proton/hydride abstraction The KDIE for AO is fully expressed on the intrinsic clearance (V max /K m ) If systemic clearance of a drug is metabolically driven by AO, pharmacokinetics could be altered

Theoretical relationship between clearance (CL) and intrinsic clearance (CL int) ) for a KDIE of 7.0 CL = Q x CL int If CL int >> Q; CL ~ = Q 1.2 07 0.7 Q + CL int Blood flow limit (Q) proto- deutero- Systemic half-life not expected to change for an IV or orally dosed drug. AUC and C max may reflect the KDIE on the extra-hepatic contributions to overall clearance CL If CL int << Q; CL = ~ Cl int 0.2 0 10 20 Systemic half-life, AUC and C max may reflect the KDIE on the Cl int -0.3 CL int /Q

Pfizer drugs examined in-vitro and in-vivo Aldehyde Oxidase component to metabolism Monoamine Oxidase component to metabolism

Intrinsic clearance KDIE for Carbazeran Substrate Human Rat Guinea Pig Cytosol Hepatocytes Cytosol Hepatocytes Cytosol S-9 Carbazeran 4.8 1.5 5.0 4.6 6.0 5.0 Metabolite profile for carbazeran 100 Intensity 80 60 40 13.12 Glucuronide m/z 537 15.23 Carbazeran m/z 361 19.54 Hydroxy Carbazeran m/z 377 Human Hepatocytes Human S9 + cofactors 20 0 100 80 Intensity 60 40 12.27 13.71 15.76 17.38 18.80 21.30 22.51 24.08 25.06 19.53 15.23 Guinea Pig S9 + cofactors 20 0 100 Inten nsity 80 60 40 20 0 13.11 13.54 15.97 16.83 20.11 23.13 23.98 15.13 Rat Hepatocytes Rat S9 + cofactors 19.57 12.96 14.40 16.39 17.34 24.82 12 14 16 18 20 22 24 Time (min)

Prediction of Carbazeran pharmacokinetic outcome from in-vitro assays In human: o PK enhancement In guinea pig and rat: o effect on systemic half-life (blood flow limited clearance ) Possible increases in Cmax and AUC due to KDIE on intrinsic i i clearance (extrahepatic AO contribution)

KDIE on PK parameters for Carbazeran (Guinea pigs) KDIE (D/H) AUC T1/2 Mean 46 4.6 08 0.8 Std. Dev 0.7 0.1 (Guinea pigs) KDIE (D/H) AUC T1/2 Cmax Mean 21.9 0.5 22.5 Std. Dev 2.6 0.1 7.7 IV-dosed Orally-dosed (Rats) KDIE (D/H) AUC T1/2 Mean 20 2.0 11 1.1 Std. Dev 0.5 0.2 (Rats) KDIE (D/H) AUC T1/2 Cmax Mean 2.3 1.27 1.5 Std. Dev. 0.2 0.1 0.0 Despite a common metabolic pathway, the guinea pig and rat differ in the outcome of KDIEs on the pharmacokinetic parameters, suggesting a species difference in their systemic clearance mechanisms

Intrinsic clearance KDIE for Zoniporide Substrate Human Rat Guinea Pig Cytosol Hepatocytes Cytosol Hepatocytes Cytosol S-9 supplemented Zoniporide 5.8 1.9 3.6 2.7 4.8 1.5 RT: 6.0-21.0 Intensity uau Metabolite profile for Zoniporide in rat hepatocytes 500000000 400000000 300000000 200000000 100000000 0 140000 120000 100000 80000 60000 40000 20000 0 13.2 M1 M10 14.44 M2 M3 15.6 16.0 16.3 M6 16.2 M5 M7 M8 M9 14.3 9.3 13.1 M4 14.6 15.9 17.1 18.8 L: 5.07E8 TIC F: + c ESI Full ms [50.00-800.00] MS data02 17.1 19.5 17.6 20.2 18.7 L: 1.57E5 nm=220.0-400.0 PDA data02 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Time (min) zoniporide

(Guinea pig) KDIE (D/H) AUC T1/2 Mean 11 1.1 12 1.2 Std. Dev 0.5 0.3 (Guinea pig) KDIE (D/H) AUC T1/2 Cmax KDIE on PK parameters for Zoniporide IV-dosed Orally-dosed (Rat) KDIE (D/H) AUC T1/2 Mean 09 0.9 14 1.4 Std. Dev 0.1 0.3 (Rat) KDIE (D/H) AUC T1/2 Cmax Mean 1.13 1.2 0.9 Mean 0.6 1.12 0.7 Std. Dev 0.1 0.2 0.1 Std. Dev 0.2 0.0 0.1 As predicted from in-vitro hepatoctyes experiments, essentially no effect is observed on the pharmacokinetics of Zoniporide in guinea pig and rat

KDIEs on the steady-state kinetic parameters for oxidation of parasubstituted phenethylamines by Monoamine Oxidase -A proteo deutero isotope effects kcat Km kcat Km D kcat D (kcat/km) substituent (min-1) ( M) (min-1) ( M) D CL int H 64 1250 7.55 1410 8.5 9.6 OH 83.7 423 35.8 633 2.3 3.5 CF3 4.5 755 0.71 1190 6.3 9.9 F 111.5 1060 21.1 2050 5.3 10.2 Cl 26.6 320 3.45 380 7.7 9.2 Br 17.1 226 2.06 260 8.3 9.6 Me 18.6 150 2.05 152 9.1 9.2 O2 200 4820 38.3 8270 5.2 9.0 Taken from: andigama and Edmondson, Biochemistry (2000), 39(49), 15258-15265

Proposed Mechanism of MAO-catalyzed deaminations

Properties and clinical results for CP409092 P lasma Concent tration (ng/ml) of (1) (D 2 )H 2 C H CP-409092 O 1000 100 10 1 O 10 mg 30mg 100mg 300mg 1000mg 0.1 0 10 20 30 40 Time (hr) Known attributes: MAO-A substrate Un-desirable clinical PK characterized by Clp/F> 200mL/min/kg; Plasma elimination t 1/2 : ~ 8.5 h Large variability in PK on-linear PK ast(ng*hr/m ml) (ng*h/ml L) AUC0-tl AUC 1000 800 600 400 200 0 10 100 1000 Dose (mg)

Low contribution of MAO to CP-409092 clearance in the rat Intrinsic clearance KDIE for CP409092 System H k/ D k Microsomes - ADPH 2.5 Microsomes + ADPH 1.1 Hepatocytes 1.1 Radiolabel Disposition of CP-409092 in Rat Metabolites Urine (U) Feces (F) U+F M1A 0.06 D 0.06 M1 0.74 6.47 7.21 M1B 0.04 0.62 0.66 M2 0.88 3.5 4.39 M2A 0.08 D 0.08 M4 (MAOpathway) 0.62 3.24 3.86 M5 0.02 0.22 0.24 (unchanged drug) 1.01 72.4 73.41 Total 3.45 66.5 89.9 Conclusion from in-vitro KDIE o effect on PK Solubility: > 5 mg/ml; Caco 2 : Papp AB : 1.2x 10-6 ; Papp: BA/AB: 7

IV and oral pharmacokinetic isotope effect for CP-409092in the rat 1000 Mean 100 IV CP-409092 PF-05136259 100 Oral CP-409092 PF-05136259 Conc ( n g /m L ) 10 centration (ng/ml) Plasma Conc 10 1 0 2 4 6 8 10 12 1 0 1 2 3 4 5 6 7 Time (hrs) Time (h) KDIE (IV) Rat# AUC T1/2 Mean 1.2 1.2 Std. Dev. 0.1 0.1

KDIE in human in vitro systems for CP409092 System H k/ D k Human Microsomes - ADPH 4.3 Microsomes + ADPH 3.8 Hepatocytes 5.7 rmao-a A 38 3.8 Large KDIE in human in vitro systems suggest possible PK enhacement The presence of other clearance routes (biliary/absorption) may not favor overall enhancement of pharmacokinetic parameters

Sandwich cultured hepatocyte billiary excretion model (1) in BD109 Rosuvastatin in BD109 30 140 25 120 pmol/mg protein 20 15 10 +Ca -Ca pmol/mg protein 100 80 60 40 Ca+ Ca- 5 20 0 0 5 10 15 Time(min) i 0 0 5 10 15 Time Uptake, app Cl b BEI (%) (pmol/min/ (ul/min/mg mg) protein) Rosuvastatin 1 7.7 2.7 34 CP-409092 1.5 0.77 30

Studies with PF-X KDIE in rat and human in vitro systems In Development 5X higher clearance at FIH than predicted MAO contributes tes to metabolism Known metabolic pathways System H k/ D k rmao-a 2.8 RLM + ADPH 1.09 Rat hepatocytes 1.06 Human hepatocytes 1.4 O X H R (D 2 )H 2 Ph H R (D 2 )H 2 CYP Ph O X O MAO-A OH O HO Ph O X Conclusion: o KDIE on the intrinsic clearance in rat hepatocytes and a small KDIE in human hepatocytes suggests deuteration will not enhance pharmacokinetics in either rat or humans

Rat IV and oral PK profiles for PF-X Conc (ng/ml) 10000 PF-04455242 IV PF-05094041 1000 100 10 1 0.1 0 4 8 12 16 20 24 Time (hrs) Plasma Con nc. of PF-4455242 (ng/ml L) Mean 1000 Oral PF-04455242 PF-04094041 100 10 1 0 4 8 12 Time (hrs) Essentially no KDIE on IV or oral PK parameters for PF-X

Considerations for deuteration as a PK enhancement strategy The identity of enzymes involved in the metabolic clearance Knowledge of their reaction mechanisms The extent of their contribution to the overall metabolic clearance Magnitude of the intrinsic i i clearance isotope effect tin hepatocytes (or equivalent) for multiple species Knowledge of other non-metabolic clearance mechanisms and the extent of their contribution to systemic clearance