Download as pdf or txt
Download as pdf or txt
You are on page 1of 33

Physiologically Based Pharmacokinetic Modeling: An Introduction

Hugh A. Barton US Environmental Protection Agency


National Center for Computational Toxicology Research Triangle Park, NC barton.hugh@epa.gov

Outline
What is pharmacokinetics (PK)? Why PK? PK Modeling Methods PBPK Model Components PBPK Model Structures

This presentation does not represent official US EPA policy.

Abbreviations
AUC: area under the concentration-time curve (mg/L*hr) Cl or Cld: clearance (L/hr) Cmax: maximum concentration (mg/L or M) PD: pharmacodynamics PK: pharmacokinetic TD: toxicodynamics TK: toxicokinetics

V or Vd: volume of distribution (L) or tissue (L) ka: absorption rate constant (conversion of volume to (1/hr) mass assumes 1 g/mL, the Km: Michaelis-Menten density of water) constant (M) Q: blood flow Vmax: maximal metabolism rate

What is Pharmacokinetics?
What the body does to a chemical Absorption, Distribution, Metabolism, Excretion (ADME) Kinetics: rates of change, PK: chemical concentrations as a function of time Pharmacokinetics = Toxicokinetics Pharmacodynamics: What the chemical does to the body (PD=TD)

Why Use PK-based Analyses?


Improved candidate selection during chemical or drug development
Cross-species comparisons of metabolism or absorption Duration of action of different formulations

Improved toxicity study design


Dose, species, dosing interval selection

Improved toxicity study interpretation


Cross-species comparisons of metabolites or tissue distribution Links to pharmacodynamics and effects

Improved risk and safety assessments


Understanding interspecies, dose, route-to-route extrapolations Evaluating population variability
Modeling populations (e.g., polymorphisms) versus individuals Modeling life stages (e.g., children, elderly, ill)

Evaluating uncertainties

Dose-response Assessment
Exposure:
external dose/concentration

Low Information Default

Pharmacokinetics:
internal tissue dose

Pharmacodynamics:
action on target tissue

Response:
measured toxicity

Which Pharmacokinetic Analysis Method?


Classical Compartmental Models Noncompartmental Models Population Pharmacokinetics PBPK Models

Classical Compartmental
10.000

Plasma (g/mL)

1.000

ka
Observed Predicted

V1 Cld V2

Cle

0.100

0.010

0.001 0 500 1000 Time (hr) 1500 2000 2500

Fits equations to data to estimate PK parameters Requires in vivo data in relevant species Can be physiological, e.g., methanol distributes with total body water, but not a requirement Limitations:
Used for nonvolatiles, though adaptable for volatiles PK changes with exposure difficult to address Limited tissue distribution characterization

Noncompartmental/Regression Analysis
Mathematical analysis of plasma time course data: trapezoid rule, nonlinear regression
Uses assumption of linear terminal phase to calculate AUC (zero to infinty) from data collected to final time point

Calculates pharmacokinetic outputs such as


AUC: area under curve CL: clearance Vss: steady state volume of distribution Tmax: time of max. concentration Cmax: max. conc.

No model structure assumptions Requires in vivo chemical concentration data in relevant species

Population Pharmacokinetics
Statistical analyses to characterize pharmacokinetic parameters for populations of people Often focuses on issues of limited data (sparse data) for each individual within the population versus extensive time course data for an individual

Physiologically Based Pharmacokinetic (PBPK) Models


PBPK models describe the organism as a set of tissue compartments interconnected by blood (plasma) flow Systems of differential equations based upon mass balance

PBPK Analysis
Captures biological processes and hypotheses explicitly
Varying degrees of detail or biological realism Flexibility to reflect biology

Can incorporate changes in PK due to chemical (e.g. GSH depletion, protein induction), growth/aging Facilitates analyses across species, doses and human population subgroups Limitations:
Greater requirements for in vitro or in vivo data Statistical evaluation of uncertainty and variability more challenging Model development and implementation requires appropriate expertise

PBPK Model Components


Model Purpose/Goal Deterministic Model
Biological Hypotheses Exposure conditions Desired outputs

Non-deterministic Model
Statistical model

Data

Model Purpose: Basic Questions


What do I need to know to carry out an analysis based upon internal dosimetry (i.e., applying pharmacokinetics)?
What toxic effects at what life stages? (i.e., potential critical studies) What species (toxicity, PK, metabolism studies)?
Toxicity testing animals Humans

What is known about the mode of action for each toxicity of interest?
Parent chemical and/or metabolite(s) (reactive or not?) Interactions with macromolecules, cells, tissues, systems? Critical for PK model structure and selection of dose metrics.

How will model be used in safety or risk assessment?


Route-to-route extrapolation (What routes?) Cross-species extrapolation Cross-chemical extrapolation

PBPK Model Components


Physiological/Anatomical
Tissue volumes Blood flow rates Cardiac output Glomerular filtration rate Alveolar ventilation rate Hematocrit Glutathione concentration DNA concentration

Biochemical/Physicochemical
Tissue:blood partition coefficient Blood:air partition coefficient Enzymatic rate constants Equilibrium or rate constants for protein binding Transporter rate constants

ADME

What Tissues (Compartments)?


Absorption Distribution
Storage (e.g., fat, bone, serum protein binding) Distributional kinetics (e.g., total body water)

Clearance
Metabolism Excretion (e.g., urine, bile, hair)

Target Tissues for Toxicity or surrogate (often blood)

Description for a Single Well-mixed Tissue Compartment


TERMS

QtCvt
Vt; At; Pt TISSUE

QtCart

Qt = tissue blood flow Cvt = venous blood concentration Pt = tissue blood partition coefficient Vt = volume of tissue At = amount of chemical in tissue

mass-balance equation:

dA t dC t = Vt = Q t C art Q t C vt dt dt C venous equilibration assumption Cvt = t Pt

Cvt: free concentration in tissue available for clearance(s)

Diffusion Limited Distribution


TISSUE Vt; At; Pt
Interstitial Fluid: 13 L
Intracellular Fluid: 33 L

Capillary Wall Blood: 2 L RBC 3 L plasma

QtCvt

PAT

Vtb; Atb

QtCart

TISSUE BLOOD
TISSUE BLOOD mass-balance equation:

PAT: permeability area


cross-product for tissue (L/hr)

dA tb dC tb = V tb = Q t (C art C vt ) + PA t (C t / Pt C bt dt dt
TISSUE mass-balance equation:

dA t dC t = Vt = PA t (C tb C t / Pt ) dt dt

Liver Compartment
rate of change of amount in liver

rate of uptake in arterial blood

rate of loss in venous blood

- by metabolism

rate of loss

VmCvl dAl = Ql (Ca Cvl ) dt K m + Cvl


When Cvl<<Km, if Vm/Km<<Ql (liver blood flow), then flow limited metabolism.
QL
GT EH CYP GST

http://www.ncsu.edu/crsc/reports/ftp/pdf/cr sc-tr02-17.pdf

Chemical-Specific Data
In vivo PK: chemical levels over time and doses
Single or repeated exposures Exposure (dosing) regimens: intravenous bolus or infusion, oral bolus, inhalation, dermal Serial determinations in multiple animals (e.g., tissue or blood concentrations) Repeated measures in same animal/human (e.g., serum, urine, feces, exhaled breath, closed chamber atmosphere)

In vitro or ex vivo
Partition coefficients: analysis of equilibrium chemical distribution to blood and tissues Protein binding rate or equilibrium constants In vitro metabolism (e.g., estimates of Km and Vmax) Changes in biochemistry following exposure (e.g., GSH depletion)

Examples of PBPK Models


Pharmacological Agents Volatile Organic Compounds Mixtures Vapors with Nasal Toxicity Lifestages

Dv kv Qc

Pharmacological Agents: All-Trans Retinoic Acid

Plasma Formulation Dsc Stratum Corneum Dsk Viable Epidermis Richly Perfused Slowly Perfused Fat Placenta ke

Qsk Qr Qs Qf Qpl

Clewell HJ 3rd, Andersen ME, Wills RJ, Latriano L. A physiologically based pharmacokinetic model for retinoic acid and its metabolites. J Am Acad Dermatol. 1997 Mar;36(3 Pt 2):S77-85.
= submodel Glucuronide kh 4-OXO Vmg , kmg CO2 kCO2 Vmx , kmx

Embryo Liver Qg kb Gut kr Intestine kf Feces ktc kct Ql

Qg

ko

Do

13-CIS

Key Factors in Risk Assessment for all-trans-Retinoic Acid


Species differences in metabolism rodents: oxidation to active form primates: glucuronidation to inactive form Exposure route differences in bioavailability rapid oral uptake can exceed capacity of glucuronidation pathway slow topical uptake subject to high affinity clearance Kinetic differences between isomers all-trans: rapid glucuronidation/clearance 13-cis: slow oxidation/clearance

Pharmacological Agents: Dichloroacetate


Injection
CL

Rest of Body
(Volume of Distribution) CVB

Urinary Clearance

Water soluble model for dichloroacetate (Barton et al., 1999)


QL CVL

Liver
ka

Metabolism

GI Lumen

drinking water or gavage

Pharmacological Agents: Diazepam


MU TE AD

Wide range of mathematical analyses:


Gueorguieva I, Nestorov IA, Rowland M. Fuzzy simulation of pharmacokinetic models: case study of whole body physiologically based model of diazepam. J Pharmacokinet Pharmacodyn. 2004 Jun;31(3):185-213. Gueorguieva I, Aarons L, Rowland M. Diazepam pharamacokinetics from preclinical to phase I using a Bayesian population physiologically based pharmacokinetic model with informative prior distributions in WinBUGS. J Pharmacokinet Pharmacodyn. 2006 Oct;33(5):571-94. Gueorguieva I, Nestorov IA, Rowland M. Reducing whole body physiologically based pharmacokinetic models using global sensitivity analysis: diazepam case study. J Pharmacokinet Pharmacodyn. 2006 Feb;33(1):1-27.

Venous Blood

SK HT BR
metab

SPL LI ST KI RE LU

Arterial Blood

CI

QP Lung

CX QC

Volatile Organic Compounds: Vinyl Chloride

CVF

Fat

QF CA

CVR

Rich

QR CA

CVS

QS Slow CA

CVL

QL Liver VMAX1 KM1 VMAX2 KM2 CA KZER KA

Clewell HJ, Gentry PR, Gearhart JM, Allen BC, Andersen ME. Comparison of cancer risk estimates for vinyl chloride using animal and human data with a PBPK model. Sci Total Environ. 2001 Jul 2;274(1-3):37-66.
CO 2

KCO 2

Reactive Metabolites (RISK) KFEE

KGSM

Glutathione Conjugate (RISKG) KGSM GSH KB

Tissue / DNA Adducts (RISKM)

KS

KO

Human risk estimates (per million) for lifetime exposure to 1 ppb vinyl chloride in air based on the incident of liver angiosarcoma in animal bioassays

Animal Bioassay Study Maltoni - mouse inhalation Maltoni - rat inhalation Feron - rat diet Maltoni - rat gavage

95% UCL Risk/million/ppb Males Females 1.52 3.27 5.17 2.24 3.05 1.1 8.68 15.7

See Clewell et al 2001 and references therein.

Human risk estimates (per million) for lifetime inhalation of 1 ppb vinyl chloride in air based on the incident of liver angiosarcoma in human epidemiological studies

Epidemiological Study Fox & Collier Jones et al. Simonato et al.


See Clewell et al 2001 and references therein.

95% UCL Risk/million/ppb 0.71 - 4.22 0.97 - 3.60 0.40 - 0.79

VOCs: Mixtures
Lung
Poorly Perf. Richly Perf.

Lung
Poorly Perf. Richly Perf.

Liver Metab Gut

Liver Metab Gut

Vm Cvl dAl = Ql (C a Cvl ) K m [1 + I / Ki ] + Cvl dt


Barton HA, Creech JR, Godin CS, Randall GM, Seckel CS. Chloroethylene mixtures: pharmacokinetic modeling and in vitro metabolism of vinyl chloride, trichloroethylene, and trans-1,2-dichloroethylene in rat. Toxicol Appl Pharmacol. 1995 Feb;130(2):237-47

Nasal Toxicity: Vinyl Acetate


MS Bogdanffy, R Sarangapani, DR Plowchalk, A Jarabek, and ME Andersen A biologically based risk assessment for vinyl acetate-induced cancer and noncancer inhalation toxicity Toxicol. Sci. 1999 51: 19-35

Life Stage & Species Extrapolations


Neonatal Model Maternal Model
Lung
Poorly Perfused Richly Perfused

Lung
Richly Perf. Poorly Perf.

Liver Mammary Gut Metabolism Liver

Metab

Gut

Placenta

Liver

Brain

Body

Embryo/Fetal Model

Conclusion
Model purpose Deterministic Biological Model
PK Determinants (ADME) Target Tissues Exposure Routes

Non-deterministic Model
Often statistical (likelihood based) Describes relationship between data and model

References Reviews
Barton HA. Computational pharmacokinetics during developmental windows of susceptibility. J Toxicol Environ Health A. 2005 Jun 11-25;68(1112):889-900 Barton HA, Pastoor TP, Baetcke K, Chambers JE, Diliberto J, Doerrer NG, Driver JH, Hastings CE, Iyengar S, Krieger R, Stahl B, Timchalk C. The acquisition and application of absorption, distribution, metabolism, and excretion (ADME) data in agricultural chemical safety assessments. Crit Rev Toxicol. 2006 Jan;36(1):9-35 Clewell HJ 3rd, Andersen ME, Barton HA. A consistent approach for the application of pharmacokinetic modeling in cancer and noncancer risk assessment. Environ Health Perspect. 2002 Jan;110(1):85-93. Himmelstein KJ and Lutz RJ (1979) A Review of the Applications of Physiologically Based Pharmacokinetic Modeling. J Pharmacokinet Biopharm 7(2):127-145. Krishnan K and Andersen ME (2001) Physiologically Based Pharmacokinetic Modeling in Toxicology. In Principles and Methods in Toxicology, 4th Edition, A. Wallace Hayes (Ed), Taylor & Francis, Philadelphia. pp 193241.

You might also like