Casaret and Dull Tox

Embed Size (px)

Citation preview

  • 7/23/2019 Casaret and Dull Tox

    1/51

    Toxicokinetics is the quantitative study of the movement of an exogenous chemical from its

    entry into the body, through its distribution to organs and tissues via the blood circulation, and to

    its final disposition by way of biotransformation and excretion. The basic kinetic concepts for the

    absorption, distribution, metabolism, and

    excretion of chemicals in the body system initially came from the study of drug actions or

    pharmacology; hence, this area of study is traditionally referred to as pharmacokinetics.

    Toxicokinetics represents

    extension of kinetic principles to the study of toxicology and encompasses applications ranging

    from the study of adverse drug effects to investigations on how disposition kinetics of exogenous

    chemicals derived from either natural or environmental sources (generally refer to as

    xenobiotics) govern their deleterious effects on organisms including humans. The study of

    toxicokinetics relies on mathematical description or modeling of the time course of toxicant

    disposition in the whole organism. The classic approach to describing the kinetics of drugs

    is to represent the body as a system of one or two compartments even though the compartments

    do not have exact correspondence to anatomical structures or physiologic processes. These

    empirical compartmental models are almost always developed to describe the kinetics of

    toxicants in readily accessible body fluids (mainly blood)

    or excreta (e.g., urine, stool, and breath). This approach is particularly suited for human studies,

    which typically do not afford organ or tissue data. n such applications, extravasculardistribution, which

    does not require detail elucidation, can be represented simply by lumped compartments. !n

    alternate and newer approach, physiologically based toxicokinetic modeling attempts to portray

    the body as an elaborate system of discrete tissue or organ compartments that are interconnected

    via the circulatory system. "hysiologically based models are capable of describing a chemical#s

    movements in body tissues or regions of toxicological interest. t also allows a priori predictions

    of how changes in specific physiological processes affect the disposition kinetics of the toxicant

    (e.g., changes in respiratory status on pulmonary absorption and exhalation of a volatile

    compound) and the extrapolation of the kinetic model across animal species to humans.

    t should be emphasi$ed that there is no inherent contradiction between the classic and

    physiologic approaches. The choice of modeling approach depends on the application context,

    the available

  • 7/23/2019 Casaret and Dull Tox

    2/51

    data, and the intended utility of the resultant model. %lassic compartmental model, as will be

    shown, requires assumptions that limit its application. n comparison, physiologic models can

    predict tissue concentrations; however, it requires much more data input and often the values of

    the required parameters cannot be estimated accurately

    or precisely, which introduces uncertainty in its prediction. &e begin with a description of the

    classic approach to toxicokinetic modeling, which offers an introduction to the basic kinetic

    concepts for toxicant absorption, distribution, and elimination. This will be followed by a brief

    review of the physiologic approach to toxicokinetic modeling that is intended to illustrate the

    construction and application of these elaborate models.

    CLASSIC TOXICOKINETICS

    deally, quantification of xenobiotic concentration at the site oftoxic insult or in'ury would afford

    the most direct information on exposureresponse relationship and dynamics of response over

    time. erial sampling of relevant biological tissues following dosing can be cost*prohibitive

    during in vivo studies in animals and is nearly impossible to perform in human exposure studies.

    The most accessible and simplest means of gathering information on

    absorption, distribution, metabolism, and elimination of a compound is to examine the time

    course of blood or plasma toxicant concentration over time. f one assumes that the concentration

    Figure 7-1. Compartmental toxicokinetic models.

    ymbols for one*compartment model+ ka is the first*order absorption rate

    constant, and kel is the first*order elimination rate constant. ymbols for twocompartment

    model+ ka is the first*order absorption rate constant into the

    central compartment (), k10 is the first*order elimination rate constant from

    the central compartment (), k12 and k21 are the first*order rate constants for

    distribution between central () and peripheral (-) compartment.

    of a chemical in blood or plasma is in some describable dynamic equilibrium with its

    concentrations in tissues, then changes in plasma toxicant concentration should reflect changes in

    tissue toxicant concentrations and relatively simple kinetic models can adequately describe the

  • 7/23/2019 Casaret and Dull Tox

    3/51

    behavior of that toxicant in the body system. %lassic toxicokinetic models typically consist of a

    central compartment representing blood and tissues that the toxicant has ready access and

    equilibration is achieved almost immediately following its introduction, along with one or more

    peripheral compartments that represent tissues in slowequilibration with the toxicant in blood

    (ig. /*). 0nce introduced into the central compartment, the toxicant distributes between central

    and peripheral compartments.

    1limination of the toxicant, through biotransformation and2or excretion, is usually assumed to

    occur from the central compartment, which should comprise the rapidly perfused visceral organs

    capabl of eliminating the toxicant (e.g., kidneys, lungs, and liver). The obvious advantage of

    compartmental toxicokinetic models is that they do not require information on tissue physiology

    or anatomic structure. These models are useful in predicting the toxicant concentrations in blood

    at different doses, in establishing the time course of accumulation of the toxicant, either in its

    parent form or as biotransformed products during continuous or episodic exposures, in defining

    concentration response (vs. doseresponse) relationships, and in guiding the choice of effective

    dose and design of dosing regimen in animal toxicity studies (3owland and To$er, 445).

    One-Compartment Model

    The most straightforward toxicokinetic assessment entails quantification of the blood or more

    commonly plasma concentrations of a toxicant at several time points after a bolus intravenous

    (iv) in'ection. 0ften, the data obtained fall on a straight line when they are plotted as the

    logarithms of plasma concentrations versus time; the kinetics

    of the toxicant is said to conform to a one*compartment model (ig. /*-). 6athematically, this

    means that the decline in plasma concentration over time profile follows a simple exponential

    pattern as represented by the following mathematical expressions+

    C 7 C8 9 e:kel9t (/*)

    or its logarithmic transform

    ogC 7 ogC8 : kel 9 t

    -.

  • 7/23/2019 Casaret and Dull Tox

    4/51

    elimination rate constant with dimensions of reciprocal time (e.g., min: or hr:). The constant

    -. or to eliminate 58> of the original body load. @y substituting C/C8 7 8.5 into

    1quation (/*), we obtain

    the following relationship between T/- and kel+

  • 7/23/2019 Casaret and Dull Tox

    5/51

    T/- 7 8.?444> (exactly 44.->) elimination. Thus, given the elimination

    T/- of a toxicant, the length of time it takes for near complete

    washout of a toxicant after discontinuation of its exposure

    can easily be estimated. !s will be seen in next section, the concept

    of T/- is applicable to toxicants that exhibit multi*exponential

    kinetics.

    &e can infer from the mono*exponential decline of blood or

    plasma concentration that the toxicant equilibrates very rapidly between

    blood and the various tissues relative to the rate of elimination,

    such that extravascular equilibration is achieved nearly instantaneously

    and maintained thereafter. Bepiction of the body system

    by a one*compartment model does not mean that the concentration

    of the toxicant is the same throughout the body, but it does assume

    that the changes that occur in the plasma concentration reflect proportional

    changes in tissue toxicant concentrations (ig. /*- upper,

    right panel). n other words, toxicant concentrations in tissues are

    expected to decline with the same elimination rate constant or T/-

    as in plasma.

    Two-Compartment Model

    !fter rapid iv administration of some toxicants, the semi*logarithmic

    plot of plasma concentration versus time does not yield a straight line

    but a curve that implies more than one dispositional phase (ig. /*-).

    n these instances, it takes some time for the toxicant to be taken up

    into certain tissue groupings, and to then reach an equilibration with

  • 7/23/2019 Casaret and Dull Tox

    6/51

    the concentration in plasma; hence, a multi*compartmental model

    is needed for the description of its kinetics in the body (ig. /*).

    The concept of tissue groupings with distinct uptake and equilibration

    rates of toxicant becomes apparent when we consider the

    factors that govern the uptake of a lipid*soluble, organic toxicant.

    Table /*- presents data on the volume and blood perfusion rate of

    various organs and tissues in a standard si$e human. rom these

    data and assuming reasonable partitioning ratios of a typical lipidsoluble,

    organic compound in the various tissue types, we can estimate

    the uptake equilibration half*times of the toxicant in each

    organ or tissue region during constant, continuous exposure. The results

    suggest that the tissues can be grouped into rapid*equilibrating

    visceral organs, moderately slow*equilibrating lean body tissues

    (mainly skeletal muscle), and very slow*equilibrating body fat; these

  • 7/23/2019 Casaret and Dull Tox

    7/51

    visceral organs. or example, very rapid metabolism or excretion

    at the visceral organs would limit distribution into the slow or very

    slowtissue groupings. !lso, there are times when equilibration rates

    of a toxicant into visceral organs overlaps with lean body tissues,

    such that the distribution kinetics of a toxicant into these two tissue

    groupings become indistinct with respect to the exponential

    decline of plasma concentration, in which case two instead of three

    tissue groupings may be observed. The concept of tissue groupings

    with respect to uptake or washout kinetics serves to 'ustify the

    seemingly simplistic, yet pragmatic, mathematical description of

    extravascular distribution by the classic two* or three*compartment

    models.

    Table

    "lasma concentration*time profile of a toxicant that exhibits

    multi*compartmental kinetics can be characteri$ed by multiexponential

    equations. or example, a two*compartment model can

    be represented by the following bi*exponential equation.

    C 7A 9 e:9t CB 9 e:9t (/*=)

    whereA andB are coefficients in units of toxicant concentration,

    and and are the respective exponential constants for the initial

    and terminal phases in units of reciprocal time. The initial () phase

    is often referred to as the distribution phase, and terminal () phase

    as the post*distributional or elimination phase. The lower, middle

    panel of ig. /*- shows a graphical resolution of the two exponential

    phases from the plasma concentration*time curve. t should be noted

    that the constant is the slope of the residual log linear plot and

    not the initial slope of the decline in the observed plasma toxicant

    concentration; that is, the initial rate of decline in plasma concentration

    approximates, but is not exactly equal, the rate constant in

  • 7/23/2019 Casaret and Dull Tox

    8/51

    1quation (/*=).

    t should be noted that distribution into and out of the tissues

    and elimination of the toxicant are ongoing at all times; that

    is, elimination does occur during the DdistributionE phase, and

    distribution between compartments is still ongoing during the

    DeliminationE phase. !s illustrated in ig. /*

  • 7/23/2019 Casaret and Dull Tox

    9/51

    concentration of gentamicin initially falls very quickly with a halflife

    of around - hours; a slow terminal phase does not emerge until

    serum concentration has fallen to less than 8> of initial concentration.

    The terminal half*life of serum gentamicin is in the range

    of = to / days. This protracted terminal half*life reflects the slow

    turnover of gentamicin sequestered in the kidneys. n fact, repeated

    administration of gentamicin leads to accumulation of gentamicin

    in the kidneys, which is a risk factor for its nephrotoxicity. @ecause

    of the complication arising from the interplay of distribution and

    elimination kinetics, it has been recommended that multiphasic disposition

    should be simply described as consisting of early and late or

    rapid and slow phases; mechanistic labels of distribution and elimination

    should be applied with some caution. astly, the initial phase

    may last for only a few minutes or for hours. &hether multiphasic

    kinetics becomes apparent depends to some extent on how often

    and when the early blood samples are obtained, and on the relative

    difference in the exponential rate constants between the early and

    later phases. f the early phase of decline in toxicant concentration

    is considerably more rapid than the later phase or phases, the timing

    of blood sampling becomes critical in the ability to resolving two

    or more phases of washout.

    ometimes three or even four exponential terms are needed to

    fit a curve to the plot of log C versus time. uch compounds are

    viewed as displaying characteristics of three* or four*compartment

    open models. The principles underlying such models are the same

    as those applied to the two*compartment open model, but the mathematics

    is more complex and beyond the scope of this chapter.

    Apparent Volume o !"#tr"but"on

    or a one*compartment model, a toxicant is assumed to equilibrate

    between plasma and tissues immediately following its entry into the

    systemic circulation. Thus, a consistent relationship should exist

  • 7/23/2019 Casaret and Dull Tox

    10/51

    between plasma concentration and the amount of toxicant in each

    tissue and, by extension, to the entire amount in the body or body

    burden. The apparent volume of distribution (Vd) is defined as the

    proportionality constant that relates the total amount of the toxicant

    in the body to its concentration in plasma, and typically has units

    of liters or liters per kilogram of body weight (3owland and To$er,

    445). Vd is the apparent fluid space into which an amount of toxicant

    is distributed in the body to result in a given plasma concentration.

    !s an illustration, envision the body as a tank containing an unknown

    volume () of well mixed water. f a known amount (mg) of dye

    is placed into the water, the volume of that water can be calculated

    indirectly by determining the dye concentration (mg2) that resulted

    after the dye has equilibrated in the tank; that is, by dividing the

    amount of dye added to the tank by the resultant concentration of

    the dye inwater. !nalogously, the apparent volume of distribution of

    a toxicant in the body is determined after iv bolus administration, and

    is mathematically defined as the quotient of the amount of chemical

    in the body and its plasma concentration. Vd is calculated as

    Vd 7Doseiv

    9AUCG

    8

    (/*5)

    whereDoseiv is the intravenous dose or known amount of chemical

    in body at time $ero; is the elimination rate constant; andAUCG

    8

    is the area under the toxicant concentration versus time curve from

    time $ero to infinity.AUCG

    8 is estimated by numerical methods, the

  • 7/23/2019 Casaret and Dull Tox

    11/51

    most common one being the trape$oidal rule (Hibaldi and "errier,

    4A-). The product, 9AUCG

    8 , in unit of concentration, is the theoretical

    concentration of toxicant in plasma if dynamic equilibration

    were achievable immediately after introduction of the toxicant into

    the systemic circulation. or a one*compartment model, immediate

    equilibration of the toxicant between plasma and tissues after an

    acute exposure does hold true, in which case Vd can be calculated

    by a simpler and more intuitive equation

    Vd 7Doseiv

    C8

    (/*?)

    where C8 is the concentration of toxicant in plasma at time $ero.

    C8 is determined by extrapolating the plasma disappearance curve

    after iv in'ection to the $ero time point (ig. /*-).

    or the more complex multi*compartmental models, Vd is calculated

    according to 1quation (/*5) that involves the computation

    of area under the toxicant concentration*time curve. 6oreover, the

    concept of an overall apparent volume of distribution is strictly applicable

    to the terminal exponential phase when equilibration of the

    toxicant between plasma and all tissue sites are attained. This has

    led some investigators to refer to the apparent volumes of distribution

    calculated by 1quation (/*5) as V (for a two*compartment

    model) or V$ (for a general multi*compartmental model); the subscript

    designation refers to the terminal exponential phase (Hibaldi

    and "errier, 4A-). t should also be noted that when 1quation (/*?)

    is applied to the situation of a multi*compartmental model, the resultant

    volume is the apparent volume of the central compartment,

    often times referred to as Vc. @y definition, Vc is the proportionality

    constant that relates the total amount of the toxicant in the central

    compartment to its concentration in plasma. t has limited utility;

  • 7/23/2019 Casaret and Dull Tox

    12/51

    for example, it can be used to calculate an iv dose of the toxicant to

    target an initial plasma concentration.

    Vd is appropriately called the apparent volume of distribution

    because it does not correspond to any real anatomical volumes. The

    magnitude of the Vd term is toxicant*specific and represents the extent

    of distribution of toxicant out of plasma and into extravascular

    sites (Table /* 2kg). The

    mechanisms of tissue sequestration include partitioning of a toxicant

    into tissue fat, high affinity binding to tissue proteins, trapping

    in speciali$ed organelles (e.g., pJ trapping of amine compounds in

    acidic lyso$omes), and concentrative uptake by active transporters.

    n fact, the equation below is an alternate form of 1quation (/*/),

    which features the interplay of binding to plasma and tissue proteins

  • 7/23/2019 Casaret and Dull Tox

    13/51

    in determining the partitioning of a toxicant in that only free

    or unbound drug can freely diffuse across membrane and cellular

    barriers.

    Vd 7 Vp C

    I

    fup

    fut,i

    9 Vt,i (/*A)

    wherefup is the unbound fraction of toxicant in plasma (Table /*

  • 7/23/2019 Casaret and Dull Tox

    14/51

  • 7/23/2019 Casaret and Dull Tox

    15/51

    removal of a toxicant from the body. @y definition, clearance has the

    units of flow (e.g., m2min or 2hr). n the classic compartmental

    context, clearance is portrayed as the apparent volume containing

    the toxicant that is cleared per unit of time. !fter intravenous bolus

    administration, total body clearance (Cl) is calculated by

    Cl 7Doseiv

    AUCG

    8

    (/*8)

    %learance can also be calculated if the volume of distribution and

    elimination rate constants are known; that is, Cl 7 Vd 9 kel for a

    one*compartment model and Cl 7 Vd 9 for a two*compartment

    model. t should be noted that the relationship is a mathematical

    derivation and does not imply that clearance is dependent upon the

    distribution volume (see later section for further commenting). !

    clearance of 88 m2min can be visuali$ed as having a 88 m

    of blood or plasma completely cleared of toxicant in each minute

    during circulation.

    The biological significance of total body clearance is better

    appreciated when it is recogni$ed that it is the sum of clearances by

    individual eliminating organs (i.e., organ clearances)+

    Cl 7 Clr C Clh C Clp CK K K (/*)

    where Clr depicts renal, Clh hepatic, and Clp pulmonary clearance.

    1ach organ clearance is in turn determined by blood perfusion flow

    through the organ and the fraction of toxicant in the arterial inflow

    that is irreversibly removed, i.e., the extraction fraction (Ei).Larious

    organ clearance models have been developed to provide quantitative

    description of clearance that is related to blood perfusion flow

    (Qi), free fraction in blood (fub), and intrinsic clearance (Clint,i)

    (&ilkinson, 4A/). or example, for hepatic clearance (Clh), if the

    delivery of the toxicant to its intracellular site of removal is ratelimited

  • 7/23/2019 Casaret and Dull Tox

    16/51

    by liver blood flow (Qh) and the toxicant is assumed to have

    equal, ready access to all the hepatocytes within the liver (i.e., the

    so*called well*stirred model)+

    Clh 7 Qh 9Eh 7 Qh 9fub 9 Clint,h

    fub 9 Clint,h C Qh

    (/*-)

    where Clint,h is the hepatic intrinsic clearance that embodies a combination

    of factors that determines the access of the toxicant to the

    en$ymatic sites (e.g., plasma protein binding and sinusoidal membrane

    transport) and the biochemical efficiency of the metaboli$ing

    en$ymes (e.g., Vmax

    K6

    for an en$yme obeying 6ichaelis kinetics).

    1quation (/*-) dictates that hepatic clearance of a toxicant from

    the blood is bounded by either liver blood flow or intrinsic clearance

    (i.e.,fub 9 Clint,h). Mote that whenfub 9 Clint,h is very much

    higher than Qh,Eh approaches unity (i.e., near complete extraction

    during each passage of toxicant through the hepatic sinusoid

    or high extraction) and Clh approaches Qh. "ut in another way,

    hepatic clearance cannot exceed the hepatic blood flow rate even

    if the maximum rate of metabolism in the liver is more rapid than

    the rate of hepatic blood flow, because the rate of overall hepatic

    clearance is limited by the delivery of the toxicant to the metabolic

    en$ymes in the liver via the blood. !t the other extreme, when

    fub 9 Clint,h is very much lower than Qh,Eh becomes quite small

    (i.e., low extraction) and Clh nearly equalsfub 9 Clint,h n this instance,

    the intrinsic clearance is relatively inefficient; hence, alteration

    in liver blood flow would have little, if any, influence on liver

    clearance of the toxicant. Thus, the concept of clearance is grounded

    in the physiological and biochemical mechanisms of an eliminating

    organ.

  • 7/23/2019 Casaret and Dull Tox

    17/51

    4elat"on#."p o El"m"nat"on 3al-L"e

    to Clearan*e and Volume

    1limination half*life (T/-) is probably the most easily understood

    pharmacokinetic concept and is an important parameter as it determines

    the persistence of a toxicant following discontinuation of

    exposure. !s will be seen in a later section, elimination half*life also

    governs the rate of accumulation of a toxciant in the body during

    continuous or repetitive exposure. 1limination half*life is dependent

    upon both volume of distribution and clearance. T/- can be

    calculated from Vd and Cl+

    T/- 7 8.?4< 9 Vd

    Cl

    (/*

  • 7/23/2019 Casaret and Dull Tox

    18/51

    8 after intravenous and extravascular dosing. @ecause

    intravenous dosing assures full (88>) delivery of the dose into

    the systemic circulation, the !N% ratio should equal the fraction

    of extravascular dose absorbed and reaches the systemic circulation

    in its intact form, and is called !oa"a!la!l!t# ($). n acute toxicokinetic

    studies, bioavailability can be determined by using different

    iv and non*iv doses according to the following equation, provided

    that the toxicant does not display dose*dependent or saturable

    kinetics.

    $ 7

    I

    AUCnon*iv

    Dosenon*iv

    II

    AUCiv

    Doseiv

    (/*=)

    whereAUCnon*iv,AUCiv,Dosenon*iv, andDoseiv are the respective

    area under the plasma concentration versus time curves and doses for

    non*iv and iv administration. @ioavailabilities for various chemicals

    range in value between 8 and . %omplete availability of chemical to

    the systematic circulation is demonstrated by$ 7 . &hen$ < ,

    less than 88> of the dose is able to reach the systemic circulation.

    @ecause the concept of bioavailability is 'udged by how much of

    the dose reaches the systemic circulation, it is often referred to as

    systemic availability. ystemic availability is determined by how

    well a toxicant is absorbed from its site of application and any

    intervening processes that could remove or inactivate the toxicant

    between its point of entry and the systemic circulation. pecifically,

    systemic availability of an orally administered toxicant is governed

  • 7/23/2019 Casaret and Dull Tox

    19/51

    by its absorption at the gastrointestinal barrier, metabolism within

    the intestinal mucosa, and metabolism and biliary excretion during

    its first transit through the liver. 6etabolic inactivation and excretion

    of the toxicant at the intestinal mucosa and the liver prior to its

    entry into the systemic circulation is called pre*systemic extraction

    or first*pass effect. The following equation accounts for the action

    of absorption and sequential first*pass extraction at the intestinal

    mucosa and the liver as determinants of the bioavailability of a

    toxicant taken orally+

    $ 7fg 9 ( :Em) 9 ( :Eh) (/*5)

    wherefg is the fraction of the applied dose that is released and

    absorbed across the mucosal barrier along the entire length of the gut,

    Em is the extent of loss due to metabolism within the gastrointestinal

    mucosa, andEh is the loss due to metabolism or biliary excretion

    during first*pass through the liver. Mote thatEh in this equation is

    same as the hepatic extractionEh defined in 1quation (/*-), which

    refers to hepatic extraction of a toxicant during recirculation. This

    means that poor oral bioavailability of a chemical can be attributed to

    multiple factors. The chemical may be absorbed to a limited extent

    because of lowaqueous solubility preventing its effective dissolution

    in the gastrointestinal fluid or poor permeability across the brushborder

    membrane of the intestinal mucosa. 1xtensive degradation by

    metabolic en$ymes residing at the intestinal mucosa and the liver

    may also hinder entry of the chemical in its intact form into the

    systemic circulation.

    The rate of absorption of a toxicant via an extravascular route

    of entry is another critical determinant of outcome, particularly in

    acute exposure situations. !s shown in ig. /*5, slowing the absorption

    rate of a toxicant, while maintaining the same extent of

    absorption or bioavailability, leads to a delay in time to peak plasma

    concentration (Tp) and a decrease in the maximum concentration

  • 7/23/2019 Casaret and Dull Tox

    20/51

    (Cmax) (compare case - to case ). The converse is true; accelerating

    absorption shortens Tp and increases Cmax. The dependence

    of Tp and Cmax on absorption rate has obvious implication in the

    speed of onset and maximum toxic effects following exposure to a

    chemical. %ase < in ig. /*5 illustrates the peculiar situation when

    the absorption rate is so much slower than the elimination rate (e.g.,

    ka I kel for a one*compartment model). The terminal rate of decline

    in plasma concentration reflects the absorption rate constant,

    instead of the elimination rate constant; that is, the washout of toxicant

    is rate*limited by slow absorption until the applied dose is

    completely absorbed or removed, beyond which time the toxicant

    remaining in the body will be cleared according the elimination

    rate*constant. This means that continual absorption of a chemical

    can affect the persistence of toxic effect following an acute

    exposure, and that it is important to institute decontamination procedure

    quickly after overdose or accidental exposure to a toxicant.

    This is especially a consideration in occupational exposure via dermal

    absorption following skin contact with permeable industrial

    chemicals.

    Metabol"te K"net"*#

    The toxicity of a chemical is in some cases attributed to its biotransformation

    product(s). Jence, the formation and subsequent

    disposition kinetics of a toxic metabolite is of considerable interest.

    !s expected, the plasma concentration of a metabolite rises as

    the parent drug is transformed into the metabolite. 0nce formed,

    the metabolite is sub'ect to further metabolism to a nontoxic byproduct

    or undergoes excretion via the kidneys or bile; hence at

    some point in time, the plasma metabolite concentration peaks and

    falls thereafter. igure /*? illustrates the plasma concentration time

    course of a primary metabolite in relation to its parent compound

    under contrasting scenarios. The left panel shows the case when

  • 7/23/2019 Casaret and Dull Tox

    21/51

    the elimination rate constant of the metabolite is much greater than

  • 7/23/2019 Casaret and Dull Tox

    22/51

    first*order toxicokinetics are independent of dose. &hen plasma protein

    binding or elimination mechanisms are saturated with increasing dose, pharmacokinetic

    parameter estimates become dose*dependent. Vd may increase,

    for example, when protein binding is saturated, allowing more free toxicant

    to distribute into extravascular sites. %onversely, Vd may decrease with

    increasing dose if tissue protein binding saturates. Then, chemical may redistribute

    more freely back into plasma. &hen chemical concentrations exceed

    the capacity for biotransformation by metabolic en$ymes, overall clearance

    of the chemical decreases. These changes may or may not have effects on

    T1/2 depending upon the magnitude and direction of changes in both Vd and

    Cl.

    the overall elimination rate constant of the parent compound (i.e.,

    km I kp). The terminal decline of the metabolite parallels that of

    the parent compound; the metabolite is cleared as quickly as it is

    formed or its washout is rate*limited by conversion from the parent

    compound. The right panel shows the opposite case when the elimination

    rate constant of the metabolite is much lower than the overall

    elimination rate constant of the parent compound (i.e., km I kp).

    The slower terminal decline of the metabolite compared to the parent

    compound simply reflects a longer elimination half*life of the

    metabolite. t should also be noted that theAUCG

    8 of the metabolite

    relative to the parent compound is dependent on the partial clearance

    of the parent drug to the metabolite and clearance of the derived

    metabolite. ! biologically active metabolite assumes toxicological

    significance when it is the ma'or metabolic product and is cleared

    much less efficiently than the parent compound.

    Saturat"on To/"*o5"net"*#

    !s already mentioned, the distribution and elimination of most

    chemicals occurs by first*order processes. Nnder first*order elimination

    kinetics, the elimination rate constant, apparent volume of

  • 7/23/2019 Casaret and Dull Tox

    23/51

    distribution, clearance and half*life are expected not to change with

    increasing or decreasing dose (i.e., dose independent). !s a result, a

    semi*logarithmic display of plasma concentration versus time over

    a range of doses shows a set of parallel plots. urthermore, plasma

    concentration at a given time or theAUCG

    8 is strictly proportional

    to dose; for example, a twofold change in dose results in an exact

    twofold change in plasma concentration orAUCG

    8 . Jowever,

    for some toxicants, as the dose of a toxicant increases, its volume

    of distribution and2or clearance may change, as shown in ig. /*/.

    This is generally referred to as non*linear or dose*dependent kinetics.

    @iotransformation, active transport processes, and protein binding

    have finite capacities and can be saturated. or instance, most

    metabolic en$ymes operate in accordance to 6ichaelis6enten kinetics

    (Hibaldi and "errier, 4A-). !s the dose is escalated and

    concentration of a toxicant at the site of metabolism approaches or

    exceeds theK6 (substrate concentration at one*half Vmax, the maximum

    metabolic capacity), the increase in rate of metabolism becomes

    less than proportional to the dose and eventually approaches a

    maximum at exceedingly high doses. The transition from first*order

    to saturation kinetics is important in toxicology because it can lead

    to prolonged persistence of a compound in the body after an acute

    exposure and excessive accumulation during repeated exposures.

    ome of the salient characteristics of nonlinear kinetics include the

    following+ () the decline in the levels of the chemical in the body is

    not exponential; (-)AUCG

    8 is not proportional to the dose; (

  • 7/23/2019 Casaret and Dull Tox

    24/51

    change in response with an increasing dose, starting at the dose at

    which saturation effects become evident.

    nhaled methanol provides an example of a chemical whose

    metabolic clearance changes from first*order kinetics at low level

    exposures to $ero*order kinetics at near toxic levels (@urbacher et al.,

    -88=). igure /*A shows predicted blood methanol concentrationtime

    profiles in female monkeys followed a -*hour controlled exposure

    in an inhalation chamber at two levels of methanol vapor,

    -88 ppm and =A88 ppm. @lood methanol kinetics at -88 ppm exposure

    follows typical first*order kinetics. !t =A88 ppm, methanol

    metabolism is fully saturated, such that the initial decline in blood

    methanol following the -*hour exposure occurs at a constant rate

    (i.e., a fixed decrease in concentration per unit time independent

    of blood concentration), rather than a constant fractional rate. !s

    a result, a rectilinear plot of blood methanol concentration versus

    time yields an initial linear decline, whereas a convex curve is observed

    in the semi*logarithmic plot (compare left and right panels

    of ig. /*A). n time, methanol metabolism converts to first*order kinetics

    when blood methanol concentration falls belowK6 (i.e., the

    6ichaelis constant for the dehydrogenase en$yme); at which time,

    blood methanol shows an exponential decline in the rectilinear plot

    and a linear decline in the semi*logarithmic plot. 6oreover, ig. /*A

    shows the greater than proportionate increase (i.e., > fourfold) in

    Cmax andAUCG

    8 as the methanol vapor concentration is raised from

    -88 ppm to =A88 ppm. t should be noted that a constant T/- or

    kel does not exist during the saturation regime; it varies depending

    upon the saturating methanol dose.

    n addition to the complication of dose*dependent kinetics,

    there are chemicals whose clearance kinetics changes over time (i.e.,

    time*dependent kinetics). ! common cause of time*dependent kinetics

  • 7/23/2019 Casaret and Dull Tox

    25/51

    is auto*induction of xenobiotic metaboli$ing en$ymes; that is,

    the substrate is capable of inducing its own metabolism through activation

    of gene transcription. The classic example of auto*induction

    is with the antiepileptic drug, carbama$epine. Baily administration

    of carbama$epine leads to a continual increase in clearance and

    shortening in elimination half*life over the first few weeks of therapy

    (@ertilsson et al., 4A?).

  • 7/23/2019 Casaret and Dull Tox

    26/51

    concentration is expected during continuous exposure and the time

    it takes for a toxicant to reach steady state is governed by its elimination

    half*life. t takes one half*life to reach 58>, four half*lives

    to reach 4, and seven half*lives to reach 44.-> of steady state.

    Time to attainment of steady state is not dependent on the intake rate

    of the toxicant. The left panel of ig. /*4 shows the same time to

    58> steady state at three different rates of intake; on the other hand,

    the steady*state concentration is strictly proportional to the intake

    rate. %hange in clearance of a toxicant often leads to a corresponding

    change in elimination half*life (see right panel of ig. /*4), in

    which case both the time to reach and magnitude of steady*state

    concentration are altered. The same steady*state principle applies

    to toxicants that exhibit multi*compartmental kinetics; except that,

    accumulation occurs in a multi*phasic fashion reflective of the multiple

    exponential half*lives for inter*compartmental distribution and

    elimination. Typically, the rise in plasma concentration is relatively

    rapid at the beginning, being governed by the early (distribution)

    half*life, and becomes slower at later times when the terminal (elimination)

    half*life takes hold.

    The concept of accumulation applies to intermittent exposure

    as well. igure /*8 shows a typical occupational exposure scenario

    to volatile chemicals at the workplace over the course of a

    week. &hether accumulation occurs from day to day and further

    from week to week depends on the intervals between exposure and

    the elimination half*life of the chemicals involved. or a chemical

    with relatively short half*life compared to the interval between

    work shifts and the Dexposure holidayE over the weekend, little accumulation

    is expected. n contrast, for a chemical with elimination

  • 7/23/2019 Casaret and Dull Tox

    27/51

    -

    to -= hours), progressive accumulation is expected over the successive

    workdays. &ashout of the chemical may not be complete over

    the weekend and result in a significant carry forward of body burden

    into the next week. t should also be noted that the overall exposure

    as measured by the !N% over the cycle of a week is dependent on

    the toxicant clearance.

    Con*lu#"on

    or many chemicals, blood or plasma chemical concentration versus

    time data can be adequately described by a one* or twocompartment,

    classic pharmacokinetic model when basic assumptions

    are made (e.g., instantaneous mixing within compartments and

    first*order kinetics). n some instances, more sophisticated models

    with increased numbers of compartments will be needed to describe

    blood or plasma toxicokinetic data; for example, if the chemical is

    preferentially sequestered and turns over slowly in select tissues.

    The parameters of the classic compartmental models are usually

    estimated by statistical fitting of data to the model equations using

    nonlinear regression methods. ! number of software packages are

    available for both data fitting and simulations with classic compartmental

    models; examples include&inMonlin ("harsight %orp., "alo

    !lto, %!), !!6 (!!6 nstitute, Nniversity of &ashington,

    eattle, &!), !B!"T (Nniversity of outhern %alifornia, os

    !ngeles, %!), and "O olutions (ummit 3esearch ervices, 6ontrose,

  • 7/23/2019 Casaret and Dull Tox

    28/51

    %0).

    +la#ma Con*

    Mon Tue 6ed T.ur )r" Sat Sun Mon

    T172 8 29 .r#

    T172 8 : .r#

    Figure 7-10. Simulated accumulation of plasma concentration from occupational

    exposure over te c!cle of a "ork "eek compared #et"een t"o

    industrial cemicals "it sort and long elimination alf-lives.

    hading represents the exposure period during the A*hour workday, 6onday

    through riday. ntake of the chemical into the systemic circulation is

    assumed to occur at a constant rate during exposure. 1xposure is negligible

    over the weekend. or the chemical with the short elimination half*life of

    A hours, minimal accumulation occurs from day to day over the work days.

    Mear complete washout of the chemical is observed when work resumes on

    6onday (see arrow). or the chemical with the long elimination half*life

    of -= hours, progressive accumulation is observed over the five work days.

    &ashout of the longer half*life chemical over the weekend is incomplete; a

    significant residual is carried into the next work week. @ecause of its lower

    clearance, the overall !N% of the long half*life chemical over the cycle of a

    week is higher by threefold.####################

  • 7/23/2019 Casaret and Dull Tox

    29/51

  • 7/23/2019 Casaret and Dull Tox

    30/51

    are now well within the reach of toxicologists.

    The advantages of physiologically based models compared

    with classic models are that () these models can describe the time

    course of distribution of toxicants to any organ or tissue, (-) they

    allow estimation of the effects of changing physiologic parameters

    on tissue concentrations, (

  • 7/23/2019 Casaret and Dull Tox

    31/51

  • 7/23/2019 Casaret and Dull Tox

    32/51

    not completely different. @oth contain a liver compartment because

    the hepatic metabolism of each chemical is an important element

    of its disposition. t is important to reali$e that there is no generic

    physiologic model. 6odels are simplifications of reality and should

    contain elements believed to represent the essential disposition features

    of a chemical.

    n view of the fact that physiologic modeling requires more effort

    than does classic compartmental modeling, what then accounts

    for the popularity of this approach among toxicologistsP The answer

    lies in the potential predictive power of physiologic models.

    Toxicologists are constantly faced with the issue of extrapolation in

    risk assessmentsQfrom laboratory animals to humans, from high to

    low doses, from occasional to continuous exposure, and from single

    chemicals to mixtures. @ecause the kinetic constants in physiologic

    models represent measurable biological or chemical processes, the

    resultant physiologic models have the potential for extrapolation

    from observed data to predicted scenarios.

  • 7/23/2019 Casaret and Dull Tox

    33/51

  • 7/23/2019 Casaret and Dull Tox

    34/51

    448). The conclusion is that the same model structure is capable of

    describing the chemicals# kinetics in two different species. @ecause

    the parameters underlying the model structure represent measurable

    biological and chemical determinants, the appropriate values

    for those parameters can be chosen for each species, forming the basis

    for successful interspecies extrapolation. 1ven though the same

    model structure is used for both rodents and humans, the simulated

    and the observed kinetics of both chemicals differ between rats and

    humans. The terminal half*life of both organics is longer in the human

    compared with the rat. This longer half*life for humans is due to

    the fact that clearance rates for smaller species are faster than those

    for larger ones. 1ven though the larger species breathes more air or

    pumps more blood per unit of time than does the smaller species,

    blood flows and ventilation rates per unit of body mass are greater

    for the smaller species. The smaller species has more breaths per

    minute or heartbeats per minute than does the larger species, even

    though each breath or stroke volume is smaller. The faster flows per

    unit mass result in a more efficient delivery of a chemical to organs

    responsible for elimination. Thus, a smaller species can eliminate

    the chemical faster than a larger one can. @ecause the parameters

    in physiologic models represent real, measurable values, such as

    blood flows and ventilation rates; the same model structure can resolve

    such disparate kinetic behaviors among species.

    Compartment#

    The basic unit of the physiologic model is the lumped compartment,

    which is often depicted as a box in a graphical scheme (ig. /*

  • 7/23/2019 Casaret and Dull Tox

    35/51

    kidney, or a widely distributed tissue type such as fat or skin. %ompartments

    usually consist of three individual well*mixed regions, or

    sub*compartments, that correspond to specific physiologic spaces

    or regions of the organ or tissue. These sub*compartments are+

    () the vascular space through which the compartment is perfused

    with blood, (-) the interstitial space that forms the matrix for the

    cells, and (

  • 7/23/2019 Casaret and Dull Tox

    36/51

  • 7/23/2019 Casaret and Dull Tox

    37/51

    information on organ and tissue volumes across species, @rownet al.

    (44/) is a good starting point.

    +.#"olo"* "hysiologic parameters encompass a wide variety of

    processes in biological systems. The most commonly used physiologic

    parameters are blood flows and lung ventilation. The blood

    flow rate (Qt in volume per unit time, such as m2min or 2h) to

    individual compartments must be known. !dditionally, information

    on the total blood flow rate or (ar)!a( o&tp&t (Qc) is necessary. f

    inhalation is the route for exposure to the chemical or is a route of

    elimination, the alveolar ventilation rate (Qp) also must be known.

    @lood flowrates and ventilation rates can be taken from the literature

    or can be obtained experimentally.

    "arameters for renal excretion and hepatic metabolism are another

    subset of physiologic parameters, and are required, if these

    processes are important in describing the elimination of a chemical.

    or example, glomerular filtration rate and renal tubular transport

    parameters are required to describe renal clearance. f a chemical is

    knownto be metaboli$ed via a saturable process, both Vmax (the maximum

    rate of metabolism) andK6 (the concentration of chemical at

    one*half Vmax) for each of the en$ymes involved must be obtained

    so that elimination of the chemical by metabolism can be described

    in the model. n principle, these parameters can be determined from

    in vitro metabolism studies with cultured cells, tissue homogenates,

    or cellular fractions containing the metabolic en$ymes (e.g., microsomes

    for cytochrome "=58 en$ymes and NB"*HTs), along with appropriate

    in vitro*in vivo scaling techniques (watsubo et al., 44/;

    6acHregor et al., -88; 6iners et al., -88?). !lthough there have

    been examples of remarkable success with quantitative prediction of

    in vivo metabolic clearance based on in vitro biochemical data, there

    are also notable failures. Nnfortunately, estimation of metabolic parameters

    from in vivo studies is also fraught with difficulties, especially

  • 7/23/2019 Casaret and Dull Tox

    38/51

    when multiple metabolic pathways and en$ymes are present.

    1stimation of metabolic parameters remains a challenging aspect of

    physiologically based toxicokinetic modeling.

    T.ermodnam"* Thermodynamic parameters relate the total concentration

    of a chemical in a tissue (Ct) to the concentration offree

    chemical in that tissue (Cf). Two important assumptions are that

    () total and free concentrations are in equilibrium with each other,

    and (-) only free chemical can be exchanged between the tissue

    sub*compartments (ut$ et al., 4A8). 6ost often, total concentration

    is measured experimentally; however, it is the free concentration

    that is available for passage across membrane barriers, binding to

    proteins, metabolism, or carrier*mediated transport. Larious mathematical

    expressions describe the relationship between these two

    entities. n the simplest situation, the toxicant is a freely diffusible

    water*soluble chemical that does not bind to any molecules. n this

    case, free concentration of the chemical is equal to the total concentration

    of the chemical in the tissue+ total 7 free, or Ct 7 Cf.

    The affinity of many chemicals for tissues of different composition

    varies. The extent to which a chemical partitions into a tissue is directly

    dependent on the composition of the tissue and independent

    of the concentration of the chemical. Thus, the relationship between

    free and total concentration becomes one of proportionality+ total 7

    free9partition coefficient, or Ct 7 Cf 9Pt. n this case,Pt is called

    a tissue partition coefficient, which is most often determined from

    tissue distribution studies in animals, ideally at steady*state during

    continuous intravenous infusion of the chemical. 1stimation ofPt

    based on in vitro binding studies with human or animal tissues or tissue

    fractions has proven successful in some cases (in et al., 4A-;

    6acHregor et al., -88). Onowledge of the value ofPt permits an

    indirect calculation of the free concentration of toxicant in the tissue

    or Cf 7 Ct/Pt.

  • 7/23/2019 Casaret and Dull Tox

    39/51

    Table /*= compares the partition coefficients for a number of

    toxic volatile organic chemicals. The larger values for the fat2blood

    partition coefficients compared with those for other tissues suggests

    that these chemicals distribute into fat to a greater extent than they

    distribute into other tissues. This has been observed experimentally.

    at and fatty tissues, such as bone marrow, contain higher concentrations

    of ben$ene than do tissues such as liver and blood. imilarly,

    styrene concentrations in fatty tissue are higher than styrene

    concentrations in other tissues. t should be noted that lipophilic

    organic compounds often can bind to plasma proteins and2or blood

    cell constituents, in which case the observed tissue2blood partition

    coefficients will be a function of both the tissue and blood partition

    coefficient (i.e.,Pt/Pb). Jence, partitioning or binding to blood

  • 7/23/2019 Casaret and Dull Tox

    40/51

  • 7/23/2019 Casaret and Dull Tox

    41/51

    e.g.,m2h) for the chemical and the total barrier surface area (A, in

    m-). The permeability coefficient takes into account the diffusivity

    of the specific chemical and the thickness of the cell membrane.

    C and C- are the respectivefree concentrations of the chemical in

    the originating and receiving compartments. Biffusional flux is enhanced

    when the barrier thickness is small, the barrier surface area

    is large, and a large concentration gradient exists. 6embrane transporters

    offer an additional route of entry into cells, and allow more

    effective tissue penetration for chemicals that have limited passive

    permeability. !lternately, the presence of efflux transporters at epithelial

    or endothelial barriers can limit toxicant penetration into critical

    organs, even for highly permeable toxicant (e.g., "*glycoprotein

    mediated efflux functions as part of the bloodbrain barrier). or

    both transporter*mediated influx and efflux processes, the kinetics

    is saturable and can be characteri$ed by Tmax (the maximum transport

    rate) andKT (the concentration of toxicant at one*half Tmax) for

    each of the transporters involved. n principle, kinetic parameters

    for passive permeability or carrier*mediated transport can be estimated

    from in vitro studies with cultured cell systems. Jowever, the

    predictability and applicability of such in vitro approaches for physiologic

    modeling has not been systematically evaluated (6acHregor

    et al., -88). !t this time, the transport parameters have to be estimated

    from in vivo data, which are at times difficult and carry some

    degree of uncertainty.

    There are two limiting conditions for the uptake of a toxicant

    into tissues+ perfusion*limited and diffusion*limited. !n understanding

    of the assumptions underlying the limiting conditions

    is critical because the assumptions change the way in which the

    model equations are written to describe the movement of a toxicant

    into and out of the compartment.

    +eru#"on-L"m"ted Compartment#

  • 7/23/2019 Casaret and Dull Tox

    42/51

    ! perfusion*limited compartment, alternately referred to as blood

    flow*limited or simply flow*limited compartment, describes the situation

    when permeability across the cellular or membrane barriers

    (PA) for a particular toxicant is much greater than the blood flow

    rate to the tissue (Qt), i.e.,PAI Qt. n this case, uptake of toxicant

    by tissue sub*compartments is limited by the rate at which

    the toxicant is presented to the tissue via the arterial inflow, and

    not by the rate at which the toxicant penetrates through the vascular

    endothelium, which is fairly porous in most tissues, or gains

    passage across the cell membranes. !s a result, equilibration of a

    toxicant between the blood in the tissue vasculature and the interstitial

    sub*compartment is maintained at all times, and the two subcompartments

    are usually lumped together as a single extracellular

    compartment. !n important exception to this vascular*interstitial

    equilibrium relationship is in the brain, where the capillary endothelium

    with its tight 'unctions poses a diffusional barrier between

    the vascular space and the brain interstitium. urthermore, as indicated

    in ig. /* -), cellular permeability generally does not

    limit the rate at which a molecule moves across cell membranes.

    or these molecules, flux across the cell membrane is fast compared

    with the tissue perfusion rate (PAI Qt), and the molecules

    rapidly distribute throughout the sub*compartments. n this case,

    free toxicant in the intracellular compartment is always in equilibrium

    with the extracellular compartment, and these tissue subcompartments

    can be lumped as a single compartment. uch a flowlimited

    tissue compartment is shown in ig. /*=. 6ovement into

    and out of the entire tissue compartment can be described by a single

  • 7/23/2019 Casaret and Dull Tox

    43/51

    equation.

    Vt 9 )Ct

    )t

    7 Qt 9 (Cin : Cout) (/*A)

    where Vt is the volume of the tissue compartment, Ct is the toxicant

    concentration in the compartment (Vt 9 C equals the amount

    of toxicant in the compartment), Vt 9 dCt/dt is the change in the

    amount of toxicant in the compartment with time, expressed as mass

    per unit of time, Qt is blood flow to the tissue, Cin is the toxicant

    concentration entering the compartment, and Cout is the toxicant

    Figure 7-1,. Scematic representation of a tissue compartment tat features

    #lood flo"limited uptake kinetics.

    3apid exchange of toxicant between the extracellular space (l&e) and intracellular

    space (l!-t l&e), unhindered by a significant diffusional barrier as

    symboli$ed by the dashed line, allows equilibrium to be maintained between

    the two sub*compartments at all times. n effect, a single compartment represents

    the tissue distribution of the toxicant. t is blood flow, C"n is the

    chemical concentration entering the compartment via the arterial inflow, and

    Cout is the chemical concentration leaving the c

  • 7/23/2019 Casaret and Dull Tox

    44/51

    1quation (/*A)) equals the difference in the rate of entry via arterial

    inflow and the rate of departure via venous outflow (right*hand side

    of 1quation (/*A)).

    n the perfusion*limited case, the concentration of chemical in

    the venous drainage from the tissue is equal to the free concentration

    of chemical in the tissue (i.e., Cout 7 Cf) when the chemical is not

    bound to blood constituents. !s was noted previously, Cf (or Cout)

    can be related to the total concentration of chemical in the tissue

    through a simple linear partition coefficient, Cout 7 Cf 7 Ct2Pt. n

    this case, the differential equation describing the rate of change in

    the amount of a chemical in a tissue becomes

    Vt 9 dCt/dt 7 Qt 9 RCin : Ct/PtS (/*4)

    n the event the chemical does bind to blood constituents, blood

    partitioning coefficient needs to be recogni$ed in the mass balance

    equation.

    Vt 9 dCt/dt 7 Qt 9 RCin : Ct/(Pt/Pb)S (/*-8)

    The physiologic model shown in ig. /*-, which was developed

    for volatile organic chemicals such as styrene and ben$ene,

    is a good example of a model in which all the compartments are

    described as flow*limited. Bistribution of a toxicant in all the compartments

    is described by using equations of the type noted above.

    n a flow*limited compartment, the assumption is that the concentrations

    of a toxicant in all parts of the tissue are in equilibrium. or

    this reason, the compartments are generally drawn as simple boxes

    (ig. /*-) or boxes with dashed lines that symboli$e the equilibrium

    between the intracellular and extracellular sub*compartments

    (ig. /*=). !dditionally, with a flow*limited model, estimates of

    fluxes between sub*compartments are not required to develop the

    mass balance differential equation for the compartment. Hiven the

    challenges in measuring flux across the vascular endothelium and

    cell membrane, this is a simplifying assumption that significantly

  • 7/23/2019 Casaret and Dull Tox

    45/51

    reduces the number of parameters required in the physiologic model.

    !"u#"on-L"m"ted Compartment#

    &hen uptake of a toxicant into a compartment is governed by its

    diffusion or transport across cell membrane barriers, the model is

    said to be diffusion*limited or barrier*limited. Biffusion*limited uptake

    or release occurs when the flux, or the transport of a toxicant

    across cell membranes, is slow compared with blood flow to the

    tissue. n this case, the permeability*area product is small compared

    with blood flow, i.e.,P! I Qt . The distribution of large polar

    molecules into tissue cells is likely to be limited by the rate at which

    the molecules pass through cell membranes. n contrast, entry into

    the interstitial space of the tissue through the leaky capillaries of the

    vascular space is usually rapid even for large molecules. igure /*5

    shows the structure of such a compartment. The toxicant concentrations

    in the vascular and interstitial spaces are in equilibrium and

    make up the extracellular sub*compartment, where uptake from the

    incoming blood is flow*limited. The rate of toxicant uptake across

    the cell membrane from the extracellular space into the intracellular

    space is limited by membrane permeability, and is thus diffusionlimited.

    Two mass balance differential equations are necessary to

    Figure 7-1. Scematic representation of a tissue compartment tat features

    mem#rane-limited uptake kinetics.

    "erfusion of blood into and out of the extracellular compartment is depicted

    by thick arrows. Transmembrane transport (flux) from the extracellular to

    the intracellular sub*compartment is depicted by thin double arrows. t is

    blood flow, C"n is chemical concentration entering the compartment, and

    Cout is chemical concentration leaving the compartment.

    describe the events in these two sub*compartments+

    E'tra(ell&lar Vt 9 dCt/dt 7 Qt 9 (Cin : Cout)

    :PAt 9 (Ct/Pt)CPAt 9 (Ct-/Pt-) (/*-)

    0ntra(ell&lar Vt- 9 dCt-/dt 7PAt 9 (Ct/Pt)

  • 7/23/2019 Casaret and Dull Tox

    46/51

    :PAt 9 (Ct-/Pt-) (/*--)

    Qt is blood flow, and C is the chemical concentration in entering

    blood (in), exiting blood (out), tissue extracellular space (t),

    or tissue intracellular space (t-). The subscript (t) for thePA term

    acknowledges the fact thatPA, reflecting either via passive diffusion

    or carrier*mediated processes, can differ between tissues. @oth

    equations feature fluxes or transfers across the cell membrane that

    are driven by free concentration. Jence, partition coefficients are

    needed to convert extracellular and intracellular tissue concentration

    to their corresponding free concentration. The physiologic model in

    ig. /* is composed of two diffusion*limited compartments each

    of which contain two sub*compartmentsQextracellular and intracellular

    space, and several perfusion*limited compartments.

    Spe*"al">ed Compartment#

    Lun The inclusion of a lung compartment in a physiologic model

    is an important consideration because inhalation is a common route

    of exposure to many volatile toxic chemicals. !dditionally, the lung

    compartment serves as an instructive example of the assumptions

    and simplifications that can be incorporated into physiologic models

    while maintaining the overall ob'ective of describing processes and

    compartments in biologically relevant terms. or example, although

    lung physiology and anatomy are complex, Jaggard (4-=) developed

    a simple approximation that sufficiently describes the uptake

    of many volatile chemicals by the lungs.!diagram of this simplified

    lung compartment is shown in ig. /*?. The assumptions inherent

    in this compartment description are as follows+ () ventilation is continuous,

    not cyclic; (-) conducting airways (nasal passages, larynx,

    trachea, bronchi, and bronchioles) function as inert tubes, carrying

    the vapor to the pulmonary or gas exchange region; (

  • 7/23/2019 Casaret and Dull Tox

    47/51

    from the inspired air appears in the arterial blood (i.e., there is no

    hold*up of chemical in the lung tissue and insignificant lung mass);

    and (5) vapor in the alveolar air and arterial blood within the lung

    compartment are in rapid equilibrium and are related byPb

  • 7/23/2019 Casaret and Dull Tox

    48/51

    merge in the vena cava and heart chambers to form mixed venous

    blood. n ig. /*, a blood compartment is created in which the

    input is the sum of the toxicant outflow from each compartment

    (Qt 9 Cvt). 0utflow from the blood compartment is a product of

    the blood concentration in the compartment and the total cardiac

    output (Qc 9 Cbl). The mass*balance differential equation for the

    blood compartment in ig. /* is as follows+

    Vbl 9 dCbl/dt 7 (Qbr 9 Cvbr C Qot 9 Cvot C Qk 9 Cvk C QlCvl)

    : Qc 9 Cbl, (/*-/)

    where Vbl is the volume of the blood compartment; C is concentration;

    Q is blood flow; l, r, ot, k, and l represent the blood, brain,

    other tissues, kidney, and liver compartments, respectively; and "r,

    "ot, "k, and "l represent the venous blood leaving the organs. Qc is

    the total blood flow equal to the sum of the venous blood flows from

    each organ.

    n contrast, the physiologic model in ig. /*- does not feature

    an explicit blood compartment. or simplicity, the blood volumes

    of the heart and the ma'or blood vessels that are not within organs

    are assumed to be negligible. The venous concentration of a chemical

    returning to the lungs is simply the weighted average of the

    concentrations in the venous blood emerging from the tissues.

    Cv 7 (Ql 9 Cvl 9 Qrp 9 Cvrp C Qpp 9 Cvpp C Qf 9 Cvf)/Qc

    (/*-A)

    where C is concentration; Q is blood flow; ", l, rp,pp, andf represent

    the venous blood entering the lungs, liver, richly perfused,

    poorly perfused, and fat tissue compartments, respectively; and "l,

    "rp, "pp, and "f represent the venous blood leaving the corresponding

    organs. Qc is the total blood flow equal to the sum of the blood

    flows exiting each organ.

    n the physiologic model in ig. /*-, the blood concentration

    entering each tissue compartment is the arterial concentration

  • 7/23/2019 Casaret and Dull Tox

    49/51

    (Cart) that was calculated above for the lung compartment

    (1quation (/*-=)). The decision to use one formulation as opposed

    to another to describe blood in a physiologic model depends on

    the role the blood plays in disposition and the type of application.

    f the toxicokinetics after intravenous in'ection is to be simulated

    or if binding to or metabolism by blood components is suspected,

    a separate compartment for the blood that incorporates these additional

    processes is required. ! blood compartment is obviously

    needed if the model were developed to explain a set of blood

    concentrationtime data for a toxicant. 0n the other hand, if blood

    is simply a conduit to the other compartments, as in the case for

    inhaled volatile organics shown in ig. /*-, the algebraic solution

    is acceptable.

    igure /*A shows the application of physiologic modeling in

    elucidating the disposition fate of methylmercury and its demethylated

    product, inorganic mercury following a single peroral administration

    in the growing adult rat (arris et al., 44

  • 7/23/2019 Casaret and Dull Tox

    50/51

    @, %, and B show two prominent features of methylmercurcy disposition+

    () methylmercury is rapidly demethylated to inorganic

    mercury, which is slowly eliminated from the brain and the kidneys,

    two ma'or sites of methylmercury toxicities; and (-) a significant

    portion of mercury is sequestered in hair and the ingestion of hair by

    the animal contributes to the remarkable persistence of mercury in

    the rat. This example illustrates the capability of physiologic models

    to deal with the varied and complex disposition kinetics of toxicants

    from a wide range of sources under a multitude of experimental and

    environmental exposure scenarios.

    Con*lu#"on#

    The second section provides an introduction to the simpler elements

    of physiologic models and the important assumptions that underlie

    model structures. or more detailed aspects of physiologic modeling,

    the readers can consult several in*depth and well*annotated

    reviews on physiologically based toxicokinetic models (%lewell and

    !nderson, 44=, 44?; 0#laherty, 44A; Orishnan and !nderson,

    -88; Orishnan et al., -88-; !nderson, -88

  • 7/23/2019 Casaret and Dull Tox

    51/51

    models describing biochemical interactions among xenobiotics,

    and more biologically realistic descriptions of tissues previously

    viewed as simple lumped compartments are 'ust a few of the

    more sophisticated applications. inally, physiologically based toxicokinetic

    models are beginning to be linked to biologically based

    toxicodynamic models to simulate the entire exposure dose

    response paradigm that is basic to the science of toxicology.