Biomath M263 Clinical Pharmacology

Similar documents
Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

BASIC PHARMACOKINETICS

PHA5128 Dose Optimization II Case Study I Spring 2013

General Principles of Pharmacology and Toxicology

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

Pharmacokinetics Overview

PHAR 7633 Chapter 20 Non Compartmental Analysis

Basic Concepts of TDM

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

Lippincott Questions Pharmacology

PHA Spring First Exam. 8 Aminoglycosides (5 points)

Multiple IV Bolus Dose Administration

USES OF PHARMACOKINETICS

Understand the physiological determinants of extent and rate of absorption

General Principles of Pharmacology and Toxicology

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

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

Pharmacokinetics PCTH 325. Dr. Shabbits September 12, C t = C 0 e -kt. Learning Objectives

Basic Pharmacokinetic Principles Stephen P. Roush, Pharm.D. Clinical Coordinator, Department of Pharmacy

Section 5.2: Pharmacokinetic properties

BIOPHARMACEUTICS and CLINICAL PHARMACY

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?

Principles of Toxicokinetics/Toxicodynanics

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

Nonlinear Pharmacokinetics

PHA Final Exam Fall 2006

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

One-Compartment Open Model: Intravenous Bolus Administration:

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

Noncompartmental Analysis (NCA) in PK, PK-based Design

The general Concepts of Pharmacokinetics

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

Effects of Liver Disease on Pharmacokinetics

NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES S3A

Nontraditional PharmD Program PRDO 7700 Pharmacokinetics Review Self-Assessment

Click to edit Master title style

Pharmacokinetic parameters: Halflife

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

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

CLINICAL PHARMACOKINETICS INDEPENDENT LEARNING MODULE

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

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

PKPD modelling to optimize dose-escalation trials in Oncology

Modeling & Simulation to support evaluation of Safety and Efficacy of Drugs in Older Patients

Human ADME Study Design Considerations in Healthy Subjects and in Patients

How to measure what happens in pharmacokinetics

Clinical Pharmacology. Pharmacodynamics the next step. Nick Holford Dept Pharmacology & Clinical Pharmacology University of Auckland, New Zealand

Pharmacokinetics of drug infusions

PHARMACOLOGY-1 PHL-313. Ali Alhoshani Office: 2B 84

Implementing receptor theory in PK-PD modeling

Modeling and Simulation to Support Development and Approval of Complex Products

Tracer studies in GSK for Discovery and Development

ICU Volume 11 - Issue 3 - Autumn Series

PHA Final Exam Fall 2001

ZIN EN ONZIN VAN ANTIBIOTICASPIEGELS BIJ NEONATEN

PHARMACOKINETICS SMALL GROUP I:

Monica Edholm Medica Medic l a Pr oducts Agency

Mechanisms of Drug Action

Sponsor / Company: Sanofi Drug substance: SAR236553/REGN727 (alirocumab)

Flecainide pharmacokinetics in healthy volunteers: the influence of urinary ph

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

Basic pharmacokinetics. Frédérique Servin APHP hôpital Bichat Paris, FRANCE

PHA First Exam Fall 2003

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

1. Immediate 2. Delayed 3. Cumulative

PHARMACOKINETICS OF DRUG ABSORPTION

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

SYNOPSIS. The study results and synopsis are supplied for informational purposes only.

Counties in the top and bottom two quintiles of both diabetes and obesity, Age-adjusted percentage of adults aged 20 years who are obese, 2007

Clinical Trials A Practical Guide to Design, Analysis, and Reporting

Adjusting phenytoin dosage in complex patients: how to win friends and influence patient outcomes

PART 1. Pharmacology

A population pharmacokinetic model for rifampicin auto induction

COMPARTMENTAL ANALYSIS OF DRUG DISTRIBUTION Juan J.L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program September 23, 2010

REVISITING THE PHARMACOKINETICS IN PATIENTS

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

Varun Goel 1, Nathalie H Gosselin 2, Claudia Jomphe 2, Husain Attarwala 1, Xiaoping (Amy) Zhang 1, JF Marier 2, Gabriel Robbie 1

Tutorial. & In case studies 1 and 2, we explore intravenous iv. & Then, we move on to extravascular dosing in case

ADAM, a hands-on patient simulator for teaching principles of drug disposition and compartmental pharmacokinetics

Clinical Study Report AI Final 28 Feb Volume: Page:

PHA5128 Dose Optimization II Case Study 3 Spring 2013

DOSE SELECTION FOR CARCINOGENICITY STUDIES OF PHARMACEUTICALS *)

Product: Omecamtiv Mecarbil Clinical Study Report: Date: 02 April 2014 Page 1

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

Case Study in Placebo Modeling and Its Effect on Drug Development

NONLINEAR PHARMACOKINETICS: INTRODUCTION

Tamer Barakat. Abdul Aziz ALShamali. Abdul Aziz ALShamali

Dr. M.Mothilal Assistant professor

Pharmacokinetics of Drugs. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

Pharmacokinetics of ibuprofen in man. I. Free and total

Pharmacokinetic Calculations

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

Population Pharmacokinetics and Pharmacodynamics as a Tool in Drug Development. Leon Aarons Manchester Pharmacy School University of Manchester

PHA 5127 FINAL EXAM FALL On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Pharmacokinetics and bioavailability derived from various body fluids. Saliva samples instead of plasma samples

FDA Use of Big Data in Modeling and Simulations

What is pharmacokinetics?

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors

PHA 5128 Final Exam Spring 2004 Version A. On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Transcription:

Training Program in Translational Science Biomath M263 Clinical Pharmacology Spring 2013 www.ctsi.ucla.edu/education/training/webcasts Wednesdays 3 PM room 17-187 CHS 4/3/2013 Pharmacokinetics and Pharmacodynamics Elliot Landaw, MD, PhD 4/10/2013 Clinical Pharmacology of Narcotics Michael Ferrante, MD 4/17/2013 Biomarkers for Early Detection, and Molecular Targeting of Putative Markers, for Ovarian Cancers 4/24/2013 Studying Tissue Pharmacokinetics by PET Assessment of Tumor Response to Chemotherapy Robin Farias-Eisner, MD, PhD Caius Radu, MD 5/1/2013 Essentials of Geriatric Psychopharmacology Helen Lavretsky, MD 5/8/2013 Changes in Pharmacokinetics and Pharmocodynamics Carla Janzen, MD, PhD During Pregnancy 5/15/2013 Pharmacokinetics and Pharmacodynamics in Renal Anjay Rastogi, MD Failure 5/22/2013 Gene Therapy Approaches to Cancer Treatment Richard Koya, MD 5/29/2013 Drug Therapy in Newborns: Peri- and Postnatal HIV treatment Yvonne Bryson, MD

Pharmacokinetics & Pharmacodynamics Basic Concepts Issues in Pharmacokinetics (PK) Clearance Half-lives and Residence Times Distribution Volumes Absorption & Bioavailability Measures Pharmacodynamics (PD) Steady State Models Linking of PK & PD April 3, 2013 (E. Landaw) M263 Clinical Pharmacology

Texts Some Resources M Rowland & RN Tozer Clinical Pharmacokinetics 4 th ed., Lippincott Williams & Wilkins 2011 AJ Atkinson et al. (eds). Principles of Clinical Pharmacology, Academic Press, 3rd Edition 2012 Web sites www.cc.nih.gov/training/training/principles.html www.pharmpk.com/ (links to PK/PD resources) Journals: Clinical Pharmacology & Therapeutics (www.ascpt.org) J. Pharmacokinetics & Pharmacodynamics

Basic - Definitions Pharmacokinetics (PK) quantitative analysis of the kinetics (time course) and steady state (SS) relationships of drug What the body does to the drug ADME Absorption Distribution Metabolism Excretion Elimination

Basic - Definitions Pharmacodynamics (PD) quantitative analysis of relation of drug concentration at an effect site (C e ) to drug effect (E). What the drug does to the body Understand Dose-Effect relationships SS: C e measured plasma concentration Non-SS: may need to use PK to infer C e

Effect Site Concentrations Dosing Regimen dose, frequency, route source: A. Atkinson Concentrations Plasma, urine, tissue, Parent and metabolites EFFECTS Rx, Toxic

Steady State vs. Kinetic Studies Steady state (SS) with constant IV infusion conc. not changing with time plasma conc. C SS reflects C tissue (usually) PK (+load) determine time until ~SS SS from Repetitive dosing (oral, IM, etc.) eventually reach constant Profile SS C max = peak ; C min = trough ; average C SS

Repetitive Dosing and Profile SS Cmax Cmin source: Rowland & Tozer

Steady State vs. Kinetic Studies Many PK/PD concepts are for SS Clearance; Volume of distribution SS PD effect for given SS conc. (time to PD SS may be longer than time to plasma SS) But some studies are kinetic e.g., single oral dose or I.V. bolus Tracer kinetic studies; PET Aim may be infer SS under rep. dosing

Linear vs Nonlinear System Linear Pharmacokinetics double the dose concentration doubles AUC proportional to dose Superposition principle (example): If {I.V. bolus} C iv (t) and {oral dose} C oral (t), then {both dosing together} C(t) C iv (t) + C oral (t) holds for small enough doses (microdoses) linearity for large doses if transport, binding, and elimination remain first order

Nonlinear Kinetics Example

Linear vs Nonlinear System single kinetic study + linearity can predict response to any input, including getting to SS but for NONlinear systems: CL, V, etc. not constant; depend on C SS, Dose requires testing at different doses; models time to SS not predicted by single dose study Common nonlinearities Saturation kinetics (Michaelis-Menten) Saturable plasma protein, tissue binding Threshold effects (e.g., glucose spilling) Induction; Neuro./hormonal regulation

Importance of Experiment Design Quality & interpretation of PK/PD data depend critically on design: Dose(s), route, and form (bolus vs infusion) What to sample Plasma, urine, tissue, PET, Total vs. unbound concentrations Parent compound, metabolites PD Effect measures What times to sample in a kinetic study Train team: record what was done, not just asked

Basic Concepts Pharmacokinetics & Pharmacodynamics Issues in Pharmacokinetics (PK) Clearance Half-lives and Residence Times Distribution Volumes Absorption & Bioavailability Measures Pharmacodynamics (PD) Steady State Models Linking of PK & PD

Organ Clearance Physiology: organ clearance as SS concept Q blood flow C B_arterial Organ of elimination C B_venous Elim. Flux = Q(C B_art C B_ven ) (mass/time) E = Single pass extraction fraction: E = Elim. flux/ input flux = (C B_art C B_ven )/C B_art Clearance Elim. flux/c ref (vol/time) If use C B_art as C ref, Clearance = E Q

Organ Clearance Clearance Elim. flux/c ref Elimination (metabolism, transport) often function of unbound C u (free plasma fraction) C u = f u C (but f u not routine measurement) Clearance = E Q high E (E>0.7), CL sensitive to Q, not f u low E (E<0.3) Q transit time E CL sensitive to f u, CYP induction or inhibition but SS exposure = f u AUC not sensitive to f u

Renal Clearance (CL R ) Easiest organ CL to measure e.g. Net CL R = (urine exc. rate)/(mid-collection C) Elim. flux = filtration + secretion reabs. GFR CL creat = 120 ml plasma water/minute CL R due just to filtration = GFR f u

Total Clearance (CL T or just CL) SS Clearances add: CL = CL R + CL H + nonrenal/nonhepatic clearance Estimating CL from single dose kinetic study i.v. Dose: CL = Dose/ C(t)dt = Dose/AUC 0 Oral Dose: CL = F Oral Dose/AUC where F = fraction of dose reaching central pool (plasma + tissue in rapid equilibrium with plasma) CL oral CL/F = Oral Dose/AUC

Trapezoidal rule: C(t) Estimating AUC...... May need to fit single exponential at end to estimate tail area to Fit model of data to entire C(t). e.g., C(t) = A 1 exp(-λ 1 t) + + A n exp(-λ n t) AUC = A 1 /λ 1 + + A n /λ n t

Using Profile SS to estimate AUC Profile SS after multiple repeated doses: use area under one cycle to estimate single dose AUC single dose AUC (from 0 to ) source: Rowland & Tozer

Predicting SS Concentration #1 Constant i.v. flux infusion I (mass/time) SS plasma conc. C SS = C( ) total CL = (total Elim. Flux)/C ref Here C ref is C SS Since patient at steady state, Elim. Flux = I Therefore, C SS = I / CL

Predicting SS Concentration #2 For repetitive oral dose D every T units of time, at Profile Steady State: average C SS = (FD/T) / CL. i.e. average C SS = (D/T) / CL oral where CL oral estimated from kinetic study by CL oral = Oral Dose/AUC = CL/F

Half-lives and Residence Times 1-compartment approximation for body: Drug distributes in single, well-mixed central pool 1 st order elimination rate k (time -1 ); volume V V logc(t) C(t) = (Dose/V)exp(-kt) k t k = CL/V V = Dose/C(0) t 1/2 = 0.693/k (half-life of drug in whole body) MRT = 1/k (Mean Residence Time in body) V = total drug distribution volume = CL MRT 0....

Is this single exponential decay? 40 What s the half-life of this drug? serum concentration (nm) 30 20 10 0 0 4 8 12 16 20 24 time after IP injection (hours)

conc. on LOG scale suggests No! 100 serum concentration (nm) 10 1 initial t 1/2 0.6 hours terminal t 1/2 14 hours 0.1 0 4 8 12 16 20 24 time after IP injection (hours)

Half-lives and Residence Times multi-compartment approximation for body: Drug distributes in central + peripheral pool(s) C(t) exhibits elimination and distribution kinetics logc(t) C(t) = A 1 exp(-λ 1 t) + A 2 exp(-λ 2 t) V 1 0....... t 2 or more half-lives, but terminal half-life not always the main factor for dosing, accumulation, etc. Relative importance each half-life depends on A i /λ i

Caution: Interpreting Terminal t 1/2 Terminal t 1/2 often rate limited by elimination BUT NOT ALWAYS! counterexample: gentamicin CL cr 6 107 ml/min terminal t 1/2 in all 90 hrs renal impairment affects mainly first half-life avg C SS still (D/T)/CL oral but dosing interval T to achieve desired C max /C min trickier to compute source: Rowland & Tozer Schentag et al. JAMA 238:327-9, 1977

Mean Residence Time (MRT) MRT = mean time molecule of drug resides in body before being irreversibly eliminated Assumes linear system May be useful summary measure when there are multiple half-lives Effective (overall) half-life = 0.693 MRT

Mean Residence Time MRT estimated from a kinetic study: Measure plasma concentration C(t) after dose: MRT AUMC/AUC tc(t)dt /AUC MRT = AUMC/AUC requires no peripheral elimination no traps linear PK 0

Mean Residence Time 1-compartment model MRT = 1/k = V 1 /CL half-life = 0.693 MRT time to reach 90% SS following constant flux infusion is 2.3 MRT s = 3.3 half-lives Multi-exponential model AUMC/AUC = w 1 (1/λ 1 ) + + w n (1/λ n ) where w i (A i /λ i ) and w 1 + + w n = 1 2.3 MRT s (i.e., 3.3 effective half-lives) is time to reach at least 84% SS

Distribution Volumes Volume of Central Pool (V 1 ) V 1 = i.v. Dose/C(0) C(0) estimated by back-extrapolating from early concentrations V 1 = plasma + tissues in rapid equilibrium by time of earliest plasma sample determines (transient) peak plasma concentration following i.v. dose

multi-compartment approximation for body: Drug distributes in central + peripheral pool(s) C(t) exhibits elimination and distribution kinetics logc(t) C(t) = A 1 exp(-λ 1 t) + A 2 exp(-λ 2 t) V 1 0....... t Back-extrapolated C(0) = A 1 + A 2 V 1 = Dose/C(0)

SS Total Distribution Volume (V SS, V D or just V) Assume at SS A( ) = total amount of drug in body at SS Define V = A( ) / C SS Hypothetical volume SS mass would have to occupy to yield same concentration as C SS V = CL MRT Provides insights into distribution, permeation, tissue binding, etc. back-extrapolated C(0) from terminal decay (i.e., V extrap ) may overestimate V

source: Rowland & Tozer t 1/2 depends on CL and V

Absorption & Bioavailability source: A. Atkinson

Bioavailability Measures of extent and rate of absorption from admin. site to measurement site (latter usually central pool, i.e. plasma) i.v. administration is gold standard for complete and instantaneous absorption single oral dose: informal measures are: C peak t peak

Bioavailability formal measures F estimates extent of absorption Separate i.v. and oral studies F = (Dose iv /Dose oral ) AUC oral /AUC iv fraction of administered dose reaching plasma MAT (mean absorption time) AUMC oral /AUC oral - AUMC iv /AUC iv Absorption rate constant (compart. model) Absorption flux time course (deconvolution)

Example: Rifampicin pretreatment reduces oral digoxin bioavailability

Pharmacokinetics & Pharmacodynamics Basic Concepts Issues in Pharmacokinetics (PK) Clearance Half-lives and Residence Times Distribution Volumes Absorption & Bioavailability Measures Pharmacodynamics (PD) Steady State Models Linking of PK & PD

Dose-Effect Relationships Drug Dose C(t) PD Effect(s) PK Covariates age sex body size organ function disease other drugs genes/markers

Simplest PD Model: Binary, 2-State Graded Effect 1 0 Drug-Receptor complex Drug Drug-Receptor Complex Ligand-binding domain k 1 Effector domain Receptor k 2 Effect Effect = Maximal effect [Drug] K D + [Drug] source: Frank M. Balis (K D = k 2 /k 1 )

Graded Dose-Effect Curve 100 Maximal effect (efficacy) 80 % of Maximal Effect 60 40 20 source: Frank M. Balis 0 0 200 400 600 800 EC 50 [Drug]

Comparing Dose-Effect Curves 100 80 Drug A Drug B % of Maximal Effect 60 40 Drug C source: Frank M. Balis 20 0 Effect = Maximal effect [Drug] K D + [Drug] 1 10 100 1000 [Drug]

Empirical Pharmacodynamic Models Fixed effect model Linear model Log-linear model E max model Sigmoid E max model Effect = E 0 + S [Drug] Effect = I + S Log([Drug]) E max [Drug] H Effect = H EC50 + [Drug] H source: Frank M. Balis

Sigmoid E max PD Model Effect (%) Effect (%) 100 80 H = 5 H = 2 H = 1 100 80 60 H = 0.5 60 40 H = 0.1 40 20 EC 50 20 EC 50 0 0 20 40 60 80 100 0 [Drug] 1 10 100 source: Frank M. Balis

Concentration and Effect vs. 10 8 Time Non-Steady State Central Compartment 100 80 Conc./ Amount 6 4 Peripheral Compartment Effect 60 40 Effect [% of E MAX ] 2 Effect Compartment 20 0 0 5 10 15 20 25 0 source: Frank M. Balis Time

Hysteresis and Proteresis Loops Intensity of Drug Effect 4 3 2 1 Hysteresis Loop (Counterclockwise) Equilibration delay in plasma and effect site conc. Formation of active metabolite Receptor up-regulation Intensity of Drug Effect 4 3 2 1 Tolerance Proteresis Loop (Clockwise) Receptor tachyphylaxis 0 0 0 1 2 3 4 0 1 2 3 4 Plasma Drug Concentration source: Frank M. Balis

PK/PD Applications Drug discovery/development Scaling (cell culture animal human) Feasible dosing, drug delivery Predict and quantify inter- & intra-patient variability Regulatory issues (FDA) Basic and Clinical Sciences Understand in vivo mechanisms Quantify PK and PD study endpoints Design of clinical studies Dosing regimens Timing of samples Identify important covariates

PK/PD Applications Therapy Optimal treatment strategies Individualization of therapy Clinical monitoring (PD) or predicting (PK/PD) efficacy and toxicity endpoints Pharmacogenetics/pharmacogenomics Hereditary variations in response (PK or PD) Identification of genes or loci Genome-based drug discovery Predict efficacy and potential adverse effects