Introduction and Overview: Objectives

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

1

Introduction and overview

Objectives
Upon completion of this chapter, you will have the ability to:

· compare and contrast the terms pharmacokinetics and biopharmaceutics


· describe absorption, distribution, metabolism and excretion (ADME) processes in pharmacokinetics
· delineate differences between intravascular and extravascular administration of drugs
· explain the compartmental model concept in pharmacokinetics
· explain what is meant by the order of a reaction and how the order defines the equation determining
the rate of the reaction
· compare and contrast a first-order and a zero-order process.

1.1 Use of drugs in disease states are engaged in the process of discovering new
drugs that are safe and effective and which are
significant additions to our armamentarium for
The use of drugs to treat or ameliorate disease goes
the treatment or prevention of disease. This pro-
back to the dawn of history. Since drugs are xeno-
cess is increasingly time-consuming, expensive,
biotics, that is compounds that are foreign to the
and often frustrating.
body, they have the potential to cause harm
Here are two statistics about new drug approval:
rather than healing, especially when they are
used inappropriately or in the wrong dose for · the average time for a new drug to be approved
the individual patient being treated. What, then, is between 7 to 9 years
is the right dose? The medieval physician/alche- · the cost of introducing a new drug is approxi-
mist Paracelsus stated: ‘‘Only the dose makes a mately $700 million to $1 billion.
thing not a poison.’’ This implies: ‘‘The dose of
Steps involved in the drug development process
a drug is enough but not too much.’’ It is the
include:
objective of this text to present some tools to
allow the determination of the proper dose – a 1. The pharmacologically active molecule or
dose that will be therapeutic but not toxic in an drug entity must be synthesized, isolated or
individual patient, possessing a particular set of extracted from various possible sources (rely-
physiological characteristics. ing on the disciplines of medicinal chemistry,
At the same time that the disciplines of medi- pharmacology, and toxicology).
cine and pharmacy strive to use existing drugs in 2. The formulation of a dosage form (i.e. tablet,
the most effective manner, scientific researchers capsules, suspension, etc.) of this drug must be

1
2 B a si c P ha r m a c ok i n e t i c s

accomplished in a manner that will deliver a distribution of drugs. However, the major break-
recommended dose to the ‘‘site of action’’ or a through in developing and defining this disci-
target tissue (employing the principles of phys- pline has come since the early 1970s.
ical pharmacy and pharmaceutics).
3. A dosage regimen (dose and dosing interval)
must be established to provide an effective con- 1.2 Important definitions and
centration of a drug in the body, as determined descriptions
by physiological and therapeutic needs (utiliz-
ing pharmacokinetics and biopharmaceutics).
Pharmacokinetics
Only a successful integration of these facets will
result in successful drug therapy. For example, an ‘‘Pharmacokinetics is the study of kinetics of
analgesic drug with a high therapeutic range can absorption, distribution, metabolism and excre-
be of little use if it undergoes a rapid decomposi- tion (ADME) of drugs and their corresponding
tion in the gastrointestinal tract and/or it fails to pharmacologic, therapeutic, or toxic responses
reach the general circulation and/or it is too irri- in man and animals’’ (American Pharmaceutical
tating to be administered parenterally. Association, 1972). Applications of pharmacoki-
Therefore, the final goal in the drug develop- netics studies include:
ment process is to develop an optimal dosage form
to achieve the desired therapeutic goals. The opti- · bioavailability measurements
mal dosage form is defined as one that provides the · effects of physiological and pathological con-
maximum therapeutic effect with the least amount ditions on drug disposition and absorption
of drug and achieves the best results consistently. · dosage adjustment of drugs in disease states, if
In other words, a large number of factors play and when necessary
an important role in determining the activity of a · correlation of pharmacological responses with
drug administered through a dosage form. It is administered doses
one of the objectives of this book to describe these · evaluation of drug interactions
factors and their influence on the effectiveness of · clinical prediction: using pharmacokinetic
these drugs. parameters to individualize the drug dosing
A variety of disciplines are involved in under- regimen and thus provide the most effective
standing the events that take place during the drug therapy.
process by which a chemical entity (substance) Please note that in every case, the use must be
becomes an active drug or a therapeutic agent. preceded by observations.
1. Principles of physics, physical chemistry, and
mathematics are essential in the formulation
of an optimum dosage form. Biopharmaceutics
2. An understanding of physiology and pharma-
cology is essential in the process of screening ‘‘Biopharmaceutics is the study of the factors
for active drug and in selecting an appropriate influencing the bioavailability of a drug in man
route of administration. and animals and the use of this information to
3. Knowledge of the principles of kinetics (rate optimize pharmacological and therapeutic activ-
processes), analytical chemistry and therapeu- ity of drug products’’ (American Pharmaceutical
tics is essential in providing an effective con- Association, 1972). Examples of some factors
centration of a drug at the ‘‘site of action.’’ include:

Pharmacokinetics and biopharmaceutics are the · chemical nature of a drug (weak acid or weak
result of such a successful integration of the var- base)
ious disciplines mentioned above. · inert excipients used in the formulation of a
The first such approach was made by Teorell dosage form (e.g. diluents, binding agents, dis-
(1937), when he published his paper on integrating agents, coloring agents, etc.)
Introduct i on and o ve rvie w 3

· method of manufacture (dry granulation and/ Volume of distribution may be thought of as the
or wet granulation) apparent volume into which a given mass of drug
· physicochemical properties of drugs (pKa, par- would need to be diluted in order to give the
ticle size and size distribution, partition coeffi- observed concentration.
cient, polymorphism, etc.). For the extravascular route, the amount of drug
that reaches general circulation is the product of
Generally, the goal of biopharmaceutical studies
the bioavailable fraction (F) and the dose admin-
is to develop a dosage form that will provide
istered. Moreover,
consistent bioavailability at a desirable rate.
The importance of a consistent bioavailability
F  Dose ¼ FX0 ¼ ðAUCÞ¥0 KV ð1:2Þ
can be very well appreciated if a drug has a nar-
row therapeutic range (e.g. digoxin) where small
Equations 1.1 and 1.2 suggest that we must
variations in blood concentrations may result in
know or determine all the parameters (i.e.
toxic or subtherapeutic concentrations.
ðAUCÞ¥0 , K, V, F) for a given drug; therefore, it is
important to know the concentration of a drug in
blood (plasma or serum) and/or the amount
Relationship between the administered dose
(mass) of drug removed in urine (excretion data).
and amount of drug in the body
A typical plasma concentration versus time pro-
file (rectilinear, R.L.) following the administra-
Only that fraction of the administered dose
tion of a drug by an extravascular route is
which actually reaches the systemic circulation
presented in Fig. 1.1.
will be available to elicit a pharmacological
effect.
For an intravenous solution, the amount of drug Onset of action
that reaches general circulation is the dose
administered. Moreover The time at which the administered drug reaches
the therapeutic range and begins to produce the
Dose ¼ X0 ¼ ðAUCÞ¥0 KV ð1:1Þ
effect.

where ðAUCÞ¥0 is the area under curve of plasma


drug concentration versus time (AUC) from time Duration of action
zero to time infinity; K is the first-order elimina-
tion rate constant and V (or Vd) is the drug’s vol- The time span from the beginning of the onset of
ume of distribution. action up to the termination of action.

MTC
Concentration (µg mL–1)

Therapeutic range
MEC
Duration of
action
Termination
of action

Time (h)
Onset of action
Figure 1.1 A typical plot (rectilinear paper) of plasma concentration versus time following the administration of a drug by an
extravascular route. MTC, minimum toxic concentration; MEC, minimum effective concentration.
4 B a si c P ha r m a c ok i n e t i c s

Table 1.1 The therapeutic range of selected drugs

Drug Therapeutic use Therapeutic range

Tobramycin (Nebcin, Tobrex) Bactericidal–antibiotic 4–8 mg L1


Digoxin (Lanoxin) Congestive heart failure (CHF) 1–2 mg L1
Carbamazepine (Tegretol) Anticonvulsant 4–12 mg L1
Theophylline Bronchial asthma 10–20 mg L1

Termination of action 1.3 Sites of drug administration

The time at which the drug concentration in the


Sites of drug administration are classified into two
plasma falls below the minimum effective con-
categories:
centration (MEC).
· intravascular routes
· extravascular routes.
Therapeutic range

The plasma or serum concentration (e.g. mg mL1) Intravascular routes


range within which the drug is likely to produce the
therapeutic activity or effect. Table 1.1 provides, as Intravascular administration can be:
an example, the therapeutic range of selected drugs.
· intravenous
· intra-arterial.
Amount of drug in the urine
Important features of the intravascular
One can monitor the drug in the urine in order to route of drug administration
obtain selected pharmacokinetic parameters of a
drug as well as other useful information such as 1. There is no absorption phase.
the bioavailability of a drug. 2. There is immediate onset of action.
Figure 1.2 represents a typical urinary plot, 3. The entire administered dose is available to
regardless of the route of drug administration. produce pharmacological effects.

10
Cumulative amount of drug in urine (mg)

(X u)∞
9

0
0 10 20 30 40 50 60 70 80 90 100
Time (h)
Figure 1.2 A typical plot (rectilinear paper) of the cumulative amount of drug in urine (Xu) against time.
Introduct i on and o ve rvie w 5

· intramuscular administration (solution and


Concentration (µg mL–1)

Therapeutic suspension)
range
· subcutaneous administration (solution and
suspension)
Drug A · sublingual or buccal administration (tablet)
Drug B · rectal administration (suppository and enema)
· transdermal drug delivery systems (patch)
· inhalation (metered dose inhaler).
Time (h)
Important features of extravascular
Figure 1.3 A typical plasma concentration versus time plot
routes of drug administration
(rectilinear paper) following the administration of a dose of a
drug by an intravascular route. 1. An absorption phase is present.
2. The onset of action is determined by factors such
as formulation and type of dosage form, route
4. This route is used more often in life-threaten- of administration, physicochemical properties
ing situations. of drugs and other physiological variables.
5. Adverse reactions are difficult to reverse or 3. The entire administered dose of a drug may not
control; accuracy in calculations and adminis- always reach the general circulation (i.e. incom-
tration of drug dose, therefore, are very critical. plete absorption).
A typical plot of plasma and/or serum concentra- Figure 1.4 illustrates the importance of the
tion against time, following the administration of absorption characteristics when a drug is admin-
the dose of a drug by intravascular route, is illus- istered by an extravascular route.
trated in Fig. 1.3. In Fig. 1.4, please note the differences in the
onset of action, termination of action and the
duration of action as a consequence of the differ-
Extravascular routes of drug administration ences in the absorption characteristics of a drug
owing to formulation differences. One may
Extravascular administration can be by a number observe similar differences in the absorption char-
of routes: acteristics of a drug when it is administered via
· oral administration (tablet, capsule, suspen- different dosage forms or different extravascular
sion, etc.) routes.

MTC

Formulation A Therapeutic
Concentration (µg mL–1)

range

MEC
Elimination phase
Formulation B
Formulation C

Absorption
Time (h)
phase
Figure 1.4 A typical plot (rectilinear paper) of plasma concentration versus time following the (oral) administration of an
identical dose of a drug via identical dosage form but different formulations. MTC, minimum toxic concentration; MEC,
minimum effective concentration.
6 B a si c P ha r m a c ok i n e t i c s

1.4 Review of ADME processes Aspirin Km Salicylic acid


(acetylsalicylic Km3 Gentisic acid
(active)
acid) (inactive)
ADME is an acronym representing the pharmaco- Km1 Km2
kinetic processes of absorption, distribution, Salicyluric
Salicyl acid
metabolism, and elimination. glucuronide (inactive)
(inactive)
Figure 1.5 Metabolism of aspirin. Km, metabolic rate
Absorption constant.

Absorption is defined as the process by which a


drug proceeds from the site of administration to
propranolol HCl (Inderal) used as a non-selective
the site of measurement (usually blood, plasma or
b-antagonist: active metabolite is 4-hydroxypro-
serum).
pranolol
diazepam (Valium) used for symptomatic relief of
tension and anxiety: active metabolite is
Distribution desmethyldiazepam.

Distribution is the process of reversible transfer of


drug to and from the site of measurement (usually Elimination
blood or plasma). Any drug that leaves the site of
measurement and does not return has undergone Elimination is the irreversible loss of drug from
elimination. The rate and extent of drug distribu- the site of measurement (blood, serum,
tion is determined by: plasma). Elimination of drugs occur by one or
1. how well the tissues and/or organs are perfused both of:
with blood · metabolism
2. the binding of drug to plasma proteins and · excretion.
tissue components
3. the permeability of tissue membranes to the
drug molecule. Excretion
All these factors, in turn, are determined and con-
trolled by the physicochemical properties and Excretion is defined as the irreversible loss of a
chemical structures (i.e. presence of functional drug in a chemically unchanged or unaltered
groups) of a drug molecule. form. An example is shown in Fig. 1.6.
The two principal organs responsible for
drug elimination are the kidney and the liver.
The kidney is the primary site for removal of a
Metabolism
drug in a chemically unaltered or unchanged
form (i.e. excretion) as well as for metabolites.
Metabolism is the process of a conversion of one
The liver is the primary organ where drug
chemical species to another chemical species
metabolism occurs. The lungs, occasionally,
(Fig. 1.5).
may be an important route of elimination for
Usually, metabolites will possess little or none
substances of high vapor pressure (i.e. gaseous
of the activity of the parent drug. However, there
anesthetics, alcohol, etc.). Another potential
are exceptions. Some examples of drugs with ther-
route of drug removal is a mother’s milk.
apeutically active metabolites are:
Although not a significant route for elimina-
procainamide (Procan; Pronestyl) used as anti- tion of a drug for the mother, the drug may
dysrhythmic agent: active metabolite is N- be consumed in sufficient quantity to affect
acetyl procainamide the infant.
Introduct i on and o ve rvie w 7

Unchanged
aspirin or
aspirin
metabolite in
urine:
Ku
aspirin

Aspirin Km Kmu
Salicylic acid Salicylic acid
(acetylsalicylic
(active) Km3 Gentisic acid
acid)
Km1 Km2 (inactive) Gentisic acid
Km3u
Salicyluric
Salicyl acid (inactive) Salicyluric acid
Km2u
glucuronide
(inactive) Salicyl
Km1u
glucuronide
Figure 1.6 Renal excretion of aspirin and its metabolites. Km, metabolic rate constant.

Disposition of the drug. This direct proportionality between


the observed plasma concentration and the
Once a drug is in the systemic circulation (imme- amount of drug eliminated and the dose adminis-
diately for intravenous administration and after tered yields the term ‘‘linear pharmacokinetics’’
the absorption step in extravascular administra- (Fig. 1.7).
tion), it is distributed simultaneously to all tissues Because of the complexity of ADME processes,
including the organ responsible for its elimina- an adequate description of the observations is
tion. The distinction between elimination and sometimes possible only by assuming a simplified
distribution is often difficult. When such a dis- model; the most useful model in pharmacokinet-
tinction is either not desired or is difficult to ics is the compartment model. The body is con-
obtain, disposition is the term used. In other ceived to be composed of mathematically
words, disposition is defined as all the processes interconnected compartments.
that occur subsequent to the absorption of
the drug. Hence, by definition, the components
of the disposition phase are distribution and Compartment concept in pharmacokinetics
elimination.
The compartment concept is utilized in phar-
macokinetics when it is necessary to describe
1.5 Pharmacokinetic models the plasma concentration versus time data ade-
quately and accurately, which, in turn, permits

After administering a dose, the change in drug con-


centration in the body with time can be described
mathematically by various equations, most of
Concentrated Transfer Region of low
which incorporate exponential terms (i.e. ex solution concentration
or ex). This suggests that ADME processes are ‘‘first
order’’ in nature at therapeutic doses and, there-
fore, drug transfer in the body is possibly mediated Rate of transfer varies with the concentration
by ‘‘passive diffusion.’’ Therefore, there is a directly in the left compartment
proportional relationship between the observed Figure 1.7 The principle of passive diffusion and the rela-
plasma concentration and/or the amount of drug tionship between the rate of transfer and the administered
eliminated in the urine and the administered dose dose of a drug.
8 B a si c P ha r m a c ok i n e t i c s

us to obtain accurate estimates of selected fun- Slow distribution suggests that the distribu-
damental pharmacokinetics parameters such as tion equilibrium is attained slowly and at a
the apparent volume of drug distribution, the finite time (from several minutes to a few hours,
elimination half life and the elimination rate depending upon the nature of the administered
constant of a drug. The knowledge of these para- drug). Furthermore, it suggests that the vascula-
meters and the selection of an appropriate equa- ture, tissues and organs are not behaving in a
tion constitute the basis for the calculation of similar fashion toward this drug and, therefore,
the dosage regimen (dose and dosing interval) we consider the body to comprise two compart-
that will provide the desired plasma concentra- ments or, if necessary, more than two
tion and duration of action for an administered compartments.
drug. Highly perfused systems, such as the liver,
The selection of a compartment model solely the kidney and the blood, may be pooled
depends upon the distribution characteristics of a together in one compartment (i.e. the central
drug following its administration. The equation compartment: compartment 1); and systems
required to characterize the plasma concentra- that are not highly perfused, such as bones, car-
tion versus time data, however, depends upon tilage, fatty tissue and many others, can also be
the compartment model chosen and the route pooled together and placed in another compart-
of drug administration. The selected model ment (i.e. the tissue or peripheral compartment:
should be such that it will permit accurate predic- compartment 2). In this type of model, the rates
tions in clinical situations. As mentioned above, of drug transfer from compartment 1 to com-
the distribution characteristics of a drug play a partment 2 and back to compartment 1 will
critical role in the model selection process. become equal at a time greater than zero (from
Generally, the slower the drug distribution in several minutes to a few hours).
the body, regardless of the route of administra- It is important to recognize that the selection
tion, the greater the number of compartments of the compartment model is contingent upon
required to characterize the plasma concentra- the availability of plasma concentration versus
tion versus time data, the more complex is the time data. Therefore, the model selection process
nature of the equation employed. On the basis is highly dependent upon the following factors.
of this observation, it is, therefore, accurate to
state that if the drug is rapidly distributed follow- 1. The frequency at which plasma samples are
ing its administration, regardless of the route of collected. It is highly recommended that
administration, a one-compartment model will plasma samples are collected as early as possi-
do an adequate job of accurately and adequately ble, particularly for first couple of hours, fol-
characterizing the plasma concentration versus lowing the administration of the dose of a drug.
time data. 2. The sensitivity of the procedure employed to
The terms rapid and slow distribution refer to analyze drug concentration in plasma sam-
the time required to attain distribution equilib- ples. (Since inflections of the plasma concen-
rium for the drug in the body. The attainment of tration versus time curve in the low-
distribution equilibrium indicates that the rate of concentration regions may not be detected
transfer of drug from blood to various organs and when using assays with poor sensitivity, the
tissues and the rate of transfer of drug from vari- use of a more sensitive analytical procedure
ous tissues and organs back into the blood have will increase the probability of choosing the
become equal. Therefore, rapid distribution sim- correct compartment model.)
ply suggests that the rate of transfer of drug from 3. The physicochemical properties (e.g. the lipo-
blood to all organ and tissues and back into blood philicity) of a drug.
have become equal instantaneously, following
the administration (intra- or extravascular) of As mentioned above, only the distribution char-
the dose of a drug. Therefore, all organs and tis- acteristics of a drug play a role in the selection of
sues are behaving in similar fashion toward the the compartment model. The chosen model, as
administered drug. well as the route of drug administration, by
Introduct i on and o ve rvie w 9

comparison, will contribute to the selection of an where Cp is the plasma drug concentration at any
appropriate equation necessary to characterize time t; and (Cp)0 is the plasma drug concentration
the plasma concentration versus time data accu- at time t ¼ 0.
rately. The following illustrations and examples, Please note that there is a single phase in the
hopefully, will delineate some of the concepts concentration versus time plot and one exponen-
discussed in this section. tial term in the equation required to describe the
data. This indicates that a one-compartment
model is appropriate in this case.
Intravenous bolus administration,
one-compartment model
Intravenous bolus administration,
Figure 1.8 is a semilogarithmic (S.L.) plot of two-compartment model
plasma concentration versus time data for a drug
administered as an intravenous bolus dose. A Figure 1.9 clearly shows the existence of two
semilogarithmic plot derives its name from the phases in the concentration versus time data.
fact that a single axis (the y-axis in this case) The first phase (curvilinear portion) represents
employs logarithmic co-ordinates, while the drug distribution in the body; and only after a
other axis (the x-axis) employs linear co- finite time (indicated by a discontinuous perpen-
ordinates. The plotted curve is a straight line, dicular line) do we see a straight line. The time at
which clearly indicates the presence of a single which the concentration versus time plot begins
pharmacokinetic phase (namely, the elimination to become a straight line represents the occur-
phase.) Since the drug is administered intrave- rence of distribution equilibrium. This suggests
nously, there is no absorption phase. The straight that drug is being distributed slowly and requires
line also suggests that distribution is instanta- a two-compartment model for accurate character-
neous; thus the drug is rapidly distributed in the ization. The equation employed to characterize
body. These data can be accurately and ade- these plasma concentration versus time data will
quately described by employing the following be biexponential (contain two exponential
mono-exponential equation terms):

Cp ¼ ðCp Þ0 e  Kt ð1:3Þ Cp ¼ Ae  at þ Be  bt ð1:4Þ

Distribution or
α phase
Cp (µg mL–1)
Cp (µg mL–1)

Post-distribution or
β phase

Time (h) Time (h)


Figure 1.8 A typical plot (semilogarithmic) of plasma con- Figure 1.9 A typical semilogarithmic plot of plasma con-
centration (Cp) versus time following the administration of an centration (Cp) versus time following the administration of an
intravenous bolus dose of a drug that is rapidly distributed in intravenous bolus dose of a drug that is slowly distributed in
the body. the body.
10 B a si c P ha r m a c ok i n e t ic s

Absorption phase

Cp (µg mL–1)
Elimination phase

Time (h)
Figure 1.10 A typical semilogarithmic plot of plasma concentration (Cp) versus time following the extravascular adminis-
tration of a dose of a drug that is rapidly distributed in the body.

where A and a are parameters associated with Ka ðXa Þt¼0  Kt


drug distribution and B and b are parameters asso- Cp ¼ ½e  e  Ka t 
VðKa  KÞ
ciated with drug post-distribution phase.
Ka FX0
Please note that there are two phases in the ¼ ½e  Kt  e  Ka t  ð1:5Þ
VðKa  KÞ
concentration versus time data in Fig. 1.9 and
that an equation containing two exponential
where Ka is the first-order absorption rate con-
terms is required to describe the data. This indi-
stant, K is the first-order elimination rate con-
cates that a two-compartment model is appropri-
stant; (Xa)t=0 is the amount of absorbable drug at
ate in this case.
the absorption site present at time zero; F is the
absorbable fraction; and X0 is the administered
dose.
Please note that a one-compartment model
Extravascular administration:
will provide an accurate description since there
one-compartment model
is only one post-absorption phase; however, since
there are two phases for the plasma concentration
The plasma concentration versus time profile pre-
versus time data, a biexponential equation is
sented in Fig. 1.10 represents a one-compartment
required to describe the data accurately.
model for a drug administered extravascularly.
There are two phases in the profile: absorption
and elimination. However, the profile clearly
indicates the presence of only one phase in the Extravascular route of drug administration,
post-absorption period. Since distribution is the two-compartment model
sole property that determines the chosen com-
partment model and, since the profile contains Figure 1.11 clearly shows the presence of three
only one phase in the post-absorption period, phases in the plasma concentration versus time
these data can be described accurately and data for a drug administered by an extravascular
adequately by employing a one-compartment route. Three phases include absorption, distribu-
model. However, a biexponential equation would tion and post-distribution. Please note that in the
be needed to characterize the concentration ver- figure, there is a clear and recognizable distinction
sus time data accurately. The following equation between the distribution and post-distribution
can be employed to characterize the data: phases. Furthermore, the plasma concentration
Introduct i on and o ve rvie w 11

Types of model in pharmacokinetics


Absorption phase
There are several types of models used:
Distribution phase · one compartment
Cp (µg mL–1)

(α phase) · two compartment


Post-distribution · three compartments or higher (not often used).
phase (β phase)
A basic model for absorption and disposition

A simple pharmacokinetic model is depicted in


Figs 1.12 and 1.13. This model may apply to any
Time (h)
extravascular route of administration.
Figure 1.11 A typical semilogarithmic plot of plasma con- The model is based on mass balance consi-
centration (Cp) versus time following the extravascular admin- derations:
istration of a dose of a drug that is slowly distributed in the
body. 1. The amount (e.g. mg) of unchanged drug and/
or metabolite(s) can be measured in urine.
2. Drug and metabolite(s) in the body (blood,
versus time profile, in the post-absorption period plasma or serum) are measured in concentra-
looks identical to that for an intravenous bolus tion units (e.g. mg mL1).
two-compartment model (Fig. 1.9). These data, 3. Direct measurement of drug at the site of
therefore, can be described accurately by employ- administration is impractical; however, it can
ing a two-compartment model and the equation be assessed indirectly.
will contain three exponential terms (one for
Mass balance considerations, therefore, dictate
each phase: absorption, distribution, and post-
that, at any time t, for the extravascular route:
distribution.)
It should be stressed that these compart- FðDoseÞ ¼ absorbable amount at the
ments do not correspond to physiologically
absorption site
defined spaces (e.g. the liver is not a compart- þ amount in the body
ment). þ cumulative amount metabolized
If the chosen model does not adequately þ cumulative amount excreted
describe the observed data (plasma concentra- unchanged
tion), another model is proposed.
The model that is ultimately chosen should and for the intravascular route:
always be the simplest possible model which is
still capable of providing an adequate description Dose ¼ amount in the body
of the observed data. The kinetic properties of a þ amount metabolized
model should always be understood if the model þ cumulative amount excreted
is used for clinical predictions. unchanged:

X u (mg)
n
tio
xcre –1 )
Absorbable E (h
Drug in the
drug at the Absorption Ku
body or
absorption K a (h–1) blood K
M m (h –
site (X a) (X ) eta 1
bo )
lis
m
X m (mg) X mu (mg)

Figure 1.12 The principle of passive diffusion and the relationship between the rate of transfer and the administered dose of
a drug following the administration of a drug by an extravascular route.
12 B a si c P ha r m a c ok i n e t ic s

100

80 Mass of drug at the

Mass of drug (mg)


absorption site

60 Mass of drug in Cumulative mass of drug


the body excreted unchanged

40 Cumulative
metabolite in urine

20

0
0 2 4 6 8 10
Time (h)
Figure 1.13 Amount of drug (expressed as a fraction of administered dose) over time in each of the compartments shown in
Fig. 1.12.

Characteristics of a one-compartment model For the purpose of this textbook, the depen-
dent variable (Y) is either mass of drug in the body
1. Equilibrium between drug concentrations in
(X), mass of drug in the urine (Xu) or the concen-
different tissues or organs is obtained rapidly
tration of drug in plasma or serum (Cp or Cs,
(virtually instantaneously), following drug
respectively). For a very small time interval, there
input. Therefore, a distinction between dis-
will be a very small change in the value of Y as
tribution and elimination phases is not
follows:
possible.
2. The amount (mass) of drug distributed in dif-
ferent tissues may be different. dY Y 2  Y 1
¼ ð1:6Þ
3. Following equilibrium, changes in drug con- dt t2  t1
centration in blood (which can be sampled)
reflect changes in concentration of drug in where dY/dt is the instantaneous rate of change in
other tissues (which cannot be sampled). function Y with respect to an infinitesimal time
interval (dt).

1.6 Rate processes


Order of a process
After a drug is administered, it is subjected to a
In the equation dY/dt ¼ KYn, the numerical value
number of processes (ADME) whose rates control
(n) of the exponent of the substance (Y) undergo-
the concentration of drug in the elusive region
ing the change is the order of the process. Typical
known as ‘‘site of action.’’ These processes affect
orders and types of process encountered in sci-
the onset of action, as well as the duration and
ence include:
intensity of pharmacological response. Some
knowledge of these rate processes is, therefore, · zero order
essential for a better understanding of the observed · first order
pharmacological activity of the administered drug. · second order
Let us introduce the symbol Y as some func- · third order
tion which changes with time (t). This means Y is · reversible
a dependent variable and time (t) is an indepen- · parallel
dent variable. · consecutive.
Introduct i on and o ve rvie w 13

K0 Intercept = X0 (Dose)
X Product (b)

where X is a substance undergoing a change


K0
X X (in another location)
Slope = –K

X (mg)
where X is a substance undergoing transfer
Figure 1.14 Process of change (zero order).

Zero- and first-order processes are most useful for t=0 Time (h) t=t
pharmacokinetics.
Figure 1.15 Rectilinear graph (R.L.) of zero-order process.
X, concentration of drug; K, rate constant.

Zero-order process

Figure 1.14 shows the process of change in a zero- Equation 1.9 is similar to other linear equa-
order process. tions (i.e. y ¼ b  mx, where b is the vertical axis
The following is the derivation of the equation intercept and m is the negative slope of the line)
for a zero-order elimination process: (Fig. 1.15).

 dY
¼ K0 Y 0 ð1:7Þ
dt Applications of zero-order processes

where K0 is the zero-order rate constant and the Applications of zero-order processes include
minus sign shows negative change over time administration of a drug as an intravenous infu-
(elimination). sion, formulation and administration of a drug
Since Y0 ¼ 1, through controlled release dosage forms and
administration of drugs through transdermal
dY drug delivery systems.
¼ K0 ð1:8Þ
dt In order to apply these general zero-order
equations to the case of zero-order drug elimina-
This equation clearly indicates that Y changes tion, we will make the appropriate substitutions
at a constant rate, since K0 is a constant (the zero- for the general variable Y.
order rate constant). This means that the change For example, substitution of X (mass of drug in
in Y must be a function of factors other than the the body at time t) for Y in Eq. 1.8 yields the zero-
amount of Y present at a given time. Factors order elimination rate equation:
affecting the magnitude of this rate could include
the amount of enzymes present, light or oxygen
dX
absorbed, and so on. ¼ K0 ð1:10Þ
The integration of Eq. 1.8 yields the following: dt

Whereas, the counterpart of the integrated Eq.


Y ¼ Y 0  K0 t ð1:9Þ
1.9 is X ¼ Xt=0 – K0t, or

where Y is the amount present at time t and Y0 is


the amount present at time zero. (For example, Y0 X ¼ X0  K 0 t ð1:11Þ
could stand for (X)t=0, the mass of drug in the
body at time zero. In the case of an intravenous where Xt=0 is the amount of drug in the body at
injection, (X)t=0 would be equal to X0, the admin- time zero. (For an intravenous injection, this
istered dose.) equals the administered dose, X0.)
14 B a si c P ha r m a c ok i n e t ic s

Unit of the rate constant (K0) for zero- The following is the derivation of the equation
order elimination of drug for a first-order elimination process, since the neg-
ative sign indicates that the amount of Y is
Since dX in Eq. 1.10 has units of mass and dt has
decreasing over time.
units of time, K0 must have units of mass/time (e.g.
mg h1). This can also be seen by the integrated
Eq. 1.11: K0t ¼ X0  X. Therefore, dY
¼ KY 1 ð1:12Þ
dt
X0  X
K0 ¼ ¼ mg h  1 where Y is again the mass of a substance under-
t  t0
going a change or a transfer, and K is the first-
order elimination rate constant. However, since
First-order process
by definition Y1 ¼ Y,
Figure 1.16 shows the process of change in a first-
order process. dY
¼ KY ð1:13Þ
dt
K
X Product (b)
Equation 1.13 tells us that the rate at which Y
where X is a substance undergoing a change changes (specifically, decreases) depends on the
K
product of the rate constant (K) and the mass of
X X (in another location) the substance undergoing the change or transfer.
Upon integration of Eq. 1.13, we obtain:
where X is a substance undergoing transfer
Figure 1.16 Process of change (first order). Y ¼ Y 0 e  Kt ð1:14Þ

R.L. paper (Equation 1.14) R.L. paper (Equation 1.15)


Intercept = ln Y0

slope = –K
ln Y
Y

t=0 Time (h) t=0 Time (h)

R.L. paper (Equation 1.16)


Intercept = log (Y0)

–K
Log Y

slope =
2.303

t=0 Time (h)


Figure 1.17 One-compartment intravenous bolus injection: three plots using rectilinear (R.L.) co-ordinates. K, rate constant;
Y can stand for mass of drug in the body (X ), concentration of drug in plasma, etc.
Introduct i on and o ve rvie w 15

S. L. (Equation 1.14) We apply the general first-order equations


Intercept = Y0 above to the case of first-order drug elimination
by making the appropriate substitutions for the
general variable Y.
–K For example, substitution of X (mass of drug in
slope =
2.303 the body at time t) for Y in Eq. 1.12 yields the first-
Y

order elimination rate equation:


dX
¼ KX1 ¼ KX ð1:17Þ
dt
t=0 t=t
Time (h)
Upon integration of Eq. 1.17, we obtain:
Figure 1.18 One-compartment intravenous bolus injection:
plot using semilogarithmic (S.L.) co-ordinates. K, rate constant;
X ¼ X0 e  Kt ð1:18Þ
Y can be X or Cp.

where X0 is the dose of intravenously injected


drug (i.v. bolus), or
or

ln Y ¼ ln Y 0  Kt ð1:15Þ ln X ¼ ln X0  Kt ð1:19Þ

or or

log Y ¼ log Y 0  Kt=2:303 ð1:16Þ log X ¼ log X0  Kt=2:303 ð1:20Þ

The above three equations for a first-order pro- Unit for a first-order rate constant, K
cess may be plotted on rectilinear co-ordinates
Eq. 1.17
(Fig. 1.17).
Use of semilogarithm paper (i.e. S.L. plot): Eq. 1.14  dX
may be plotted (Y versus t) on semilogarithmic co- ¼ KX
dt
ordinates. It will yield a vertical axis intercept of Y0
and a slope of K/2.303 (Fig. 1.18). or dX/dt  X1 ¼ K, where units are mg h1 
mg1. So K has units of h1.
Applications

First-order elimination is extremely important Comparing zero- and first-order processes


in pharmacokinetics since the majority of
therapeutic drugs are eliminated by this Tables 1.2 and 1.3 compare zero-order and first-
process. order processes.

Table 1.2 Comparison of zero-order and first-order reactions

Terms Zero order First order

dX/dt ¼K0 (Eq. 1.10); rate remains constant ¼KX (Eq. 1.17); rate changes over time
rate constant ¼K0 (unit ¼ mg h1) ¼K (unit ¼ h1)
X X ¼ X0  Kt (Eq. 1.11) (integrated ln X ¼ ln X0  Kt (Eq. 1.19) or log X ¼ log X0  Kt/2.303 (Eq. 1.20)
equation) (integrated equation)
X0 Assume is 100 mg or 100% Assume is 100 mg or 100%
1
rate K0¼10 mg h K¼0.1 h1 or 10% of the remaining X
16 B a si c P ha r m a c ok i n e t ic s

Table 1.3 Values for parameters over time in zero- and first-order processes

Time (h) Zero order First order

X (mg) dX/dt (mg h1) X (mg) dX/dt (mg h1)

0 100 – 100 –
1 90 10 90 10
2 80 10 81 9
3 70 10 72.9 8.10
4 60 10 65.61 7.29
5 50 10 59.05 6.56
6 40 10 53.14 5.91
7 30 10 47.82 5.32
8 20 10 43.04 4.78
9 10 10 38.74 4.30
2
Mathematical review

Objectives
Upon completion of this chapter, you will have the ability to:

· correctly manipulate arithmetic and algebraic expressions, expressing the result in the correct
number of significant figures
· compare and contrast the terms variable, constant and parameter
· correctly manipulate the units in a calculation
· explain the interrelationship between slope, rate, and derivative
· construct sketches (profiles) illustrating pharmacokinetic equations.

2.1 Introduction conclusion, such as the proper dosage regimen


for the patient or the projected course of the
blood concentration of drug over time.
Pharmacokinetics is a mathematical subject. It
The exact, and exacting, nature of the science
deals in quantitative conclusions, such as a dose
of pharmacokinetics requires some degree of
or a concentration of drug in the blood. There is a
facility in mathematical manipulation. The
single correct numerical answer (along with
objective of this section is to refamiliarize the
many incorrect answers) for a pharmacokinetic
reader with some fundamental mathematical
problem. Therefore, pharmacokinetics meets
concepts that were learned once but, perhaps,
Lord Kelvin’s criterion (1889) for substantial sci-
forgotten.
entific knowledge: ‘‘I often say that when you can
measure what you are speaking about, and
express it in numbers, you know something about
2.2 A brief history
it, but when you cannot express it in numbers
of pharmacokinetics
your knowledge is of a meagre and unsatisfactory
kind.’’
Pharmacokinetics concerns itself with a partic- The mathematics of pharmacokinetics strongly
ular set of mathematical problems: the so-called resembles, and arises from, the mathematics of
‘‘word problems.’’ This type of problem presents chemical kinetics, enzyme kinetics, and radioiso-
additional challenges to the problem solver: tope (tracer) kinetics. Table 2.1 shows how, over
translating the words and phrases into mathe- the years, the mathematical theory of pharmaco-
matical symbols and equations, performing the kinetics and that of its older siblings has been
mathematical manipulations and finally translat- substantiated by experimental work. In fact, sub-
ing the result into a clinically meaningful stantiation of a particular kinetic theory often

17

You might also like