Practical pharmacokinetics

Published on 02/03/2015 by admin

Filed under Basic Science

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1176 times

3 Practical pharmacokinetics

Clinical pharmacokinetics may be defined as the study of the time course of the absorption, distribution, metabolism and excretion of drugs and their corresponding pharmacological response. In practice, pharmacokinetics makes it possible to model what may happen to a drug after it has been administered to a patient. Clearly, this science may be applied to a wide range of clinical situations, hence the term ‘clinical pharmacokinetics’. However, no matter how elegant or precise the mathematical modelling, the relationship between concentration and effect must be established before pharmacokinetics will be of benefit to the patient.

Application to therapeutic drug monitoring

Clinical pharmacokinetics is usually associated with therapeutic drug monitoring (TDM), and its subsequent utilisation. When TDM is used appropriately, it has been demonstrated that patients suffer fewer side effects than those who are not monitored (Reid et al., 1990). Although TDM is a proxy outcome measure, a study with aminoglycosides (Crist et al., 1987) demonstrated shorter hospital stays for patients where TDM was used. Furthermore, a study on the use of anticonvulsants (McFadyen et al., 1990) showed better epilepsy control in those patients where TDM was used. A literature review of the cost-effectiveness of TDM concluded that emphasis on just cost is inappropriate and clinical relevance should be sought (Touw et al., 2007). There are various levels of sophistication for the application of pharmacokinetics to TDM. Knowledge of the distribution time and an understanding of the concept of steady state can facilitate determination of appropriate sampling times.

For most drugs that undergo first-order elimination, a linear relationship exists between dose and concentration, which can be used for dose adjustment purposes. However, if the clearance of the drug changes as the concentration changes (e.g. phenytoin), then an understanding of the drug’s pharmacokinetics will assist in making correct dose adjustments.

More sophisticated application of pharmacokinetics involves the use of population pharmacokinetic data to produce initial dosage guidelines, for example nomograms for digoxin and gentamicin, and to predict drug levels. Pharmacokinetics can also assist in complex dosage individualisation using actual patient specific drug level data.

Given the wide range of clinical situations in which pharmacokinetics can be applied, pharmacists must have a good understanding of the subject and of how to apply it to maximise their contribution to patient care.

Basic concepts

Volume of distribution

The apparent volume of distribution (Vd) may be defined as the size of a compartment which will account for the total amount of drug in the body (A) if it were present in the same concentration as in plasma. This means that it is the apparent volume of fluid in the body which results in the measured concentration of drug in plasma (C) for a known amount of drug given, that is:

image

This relationship assumes that the drug is evenly distributed throughout the body in the same concentration as in the plasma. However, this is not the case in practice, since many drugs are present in different concentrations in various parts of the body. Thus, some drugs which concentrate in muscle tissue have a very large apparent volume of distribution, for example digoxin. This concept is better explained in Fig. 3.2.

The apparent volume of distribution may be used to determine the plasma concentration after an intravenous loading dose:

(1) image

Conversely, if the desired concentration is known, the loading dose may be determined:

(2) image

In the previous discussion, it has been assumed that after a given dose a drug is instantaneously distributed between the various tissues and plasma. In practice this is seldom the case. For practical purposes it is reasonable to generalise by referring to plasma as one compartment and tissue as if it were another single separate compartment. However, in reality there will be many tissue subcompartments. Thus, in pharmacokinetic terms the body may be described as if it were divided into two compartments: the plasma and the tissues.

Figure 3.3 depicts the disposition of a drug immediately after administration and relates this to the plasma concentration–time graph.

Initially, the plasma concentration falls rapidly, due to distribution and elimination (α phase). However, when an equilibrium is reached between the plasma and tissue (i.e. the distribution is complete), the change in plasma concentration is only due to elimination from the plasma (β phase), and the plasma concentration falls at a slower rate. The drug is said to follow a two-compartment model. However, if distribution is completed quickly (within minutes), then the α phase is not seen, and the drug is said to follow a one-compartment model.

The practical implications of a two-compartment model are that any sampling for monitoring purposes should be carried out after distribution is complete. In addition, intravenous bolus doses are given slowly to avoid transient side effects caused by high peak concentrations.

Elimination

Drugs may be eliminated from the body by a number of routes. The primary routes are excretion of the unchanged drug in the kidneys, or metabolism (usually in the liver) into a more water soluble compound for subsequent excretion in the kidneys, or a combination of both.

The main pharmacokinetic parameter describing elimination is clearance (CL). This is defined as the volume of plasma completely emptied of drug per unit time. For example, if the concentration of a drug in a patient is 1 g/L and the clearance is 1 L/h, then the rate of elimination will be 1 g/h. Thus, a relationship exists:

(3) image

Total body elimination is the sum of the metabolic rate of elimination and the renal rate of elimination. Therefore:

image

Thus, if the fraction eliminated by the renal route is known (fe), then the effect of renal impairment on total body clearance can be estimated.

The clearance of most drugs remains constant for each individual. However, it may alter in cases of drug interactions, changing end-organ function or autoinduction. Therefore, it is clear from equation (Eq.) (3) that as the plasma concentration changes so will the rate of elimination. However, when the rate of administration is equal to the rate of elimination, the plasma concentration is constant (Css) and the drug is said to be at a steady state.

At steady state:

image

At the beginning of a dosage regimen the plasma concentration is low. Therefore, the rate of elimination from Eq. (3) is less than the rate of administration, and accumulation occurs until a steady state is reached (see Fig. 3.1).

(4) image

It is clear from Eq. (3) that as the plasma concentration falls (e.g. on stopping treatment or after a single dose), the rate of elimination also falls. Therefore, the plasma concentration–time graph follows a non-linear curve characteristic of this type of first-order elimination (Fig. 3.4). This is profoundly different from a constant rate of elimination irrespective of plasma concentration, which is typical of zero-order elimination.

For drugs undergoing first-order elimination, there are two other useful pharmacokinetic parameters in addition to the volume of distribution and clearance. These are the elimination rate constant and elimination half-life.

The elimination rate constant (ke) is the fraction of the amount of drug in the body (A) eliminated per unit time. For example, if the body contains 100 mg of a drug and 10% is eliminated per unit time, then ke = 0.1. In the first unit of time, 0.1 × 100 mg or 10 mg is eliminated, leaving 90 mg. In the second unit of time, 0.1 × 90 mg or 9 mg is eliminated, leaving 81 mg. Elimination continues in this manner. Therefore:

(5) image

Combining Eqs. (3) and (5) gives

image

and since

image

then

image

Therefore,

(6) image

Elimination half-life (t1/2) is the time it takes for the plasma concentration to decay by half. In five half-lives the plasma concentration will fall to approximately zero (see Fig. 3.4).

The equation which is described in Fig. 3.4 is

(7) image

where C1 and C2 are plasma concentrations and t is time.

If half-life is substituted for time in Eq. (7), C2 must be half of C1.

Therefore,

image

image

image

image

(8) image

There are two ways of determining ke, either by estimating the half-life and applying Eq. (8) or by substituting two plasma concentrations in Eq. (7) and applying natural logarithms:

image

image

image

In the same way as it takes approximately five half-lives for the plasma concentration to decay to zero after a single dose, it takes approximately five half-lives for a drug to accumulate to the steady state on repeated dosing or during constant infusion (see Fig. 3.1).

This graph may be described by the equation

(9) image

where C is the plasma concentration at time t after the start of the infusion and Css is the steady state plasma concentration. Thus (if the appropriate pharmacokinetic parameters are known), it is possible to estimate the plasma concentration any time after a single dose or the start of a dosage regimen.

Dosing regimens

From the preceding sections, it is possible to derive equations which can be applied in clinical practice.

From Eq. (1) we can determine the change in plasma concentration ΔC immediately after a single dose:

(10) image

where F is bioavailability and S is the salt factor, which is the fraction of active drug when the dose is administered as a salt (e.g. aminophylline is 80% theophylline, therefore S = 0.8).

Conversely, to determine a loading dose:

(11) image

At the steady state it is possible to determine maintenance dose or steady state plasma concentrations from a modified Eq. (4):

(12) image

where T is the dosing interval.

Dosage adjustment

Under most circumstances, provided the preceding criteria are observed, adjusting the dose of a drug is relatively simple, since a linear relationship exists between the dose and concentration if a drug follows first-order elimination (Fig. 3.6A). This is the case for most drugs.

Capacity limited clearance

If a drug is eliminated by the liver, it is possible for the metabolic pathway to become saturated, since it is an enzymatic system. Initially the elimination is first-order, but once saturation of the system occurs, elimination becomes zero-order. This results in the characteristic dose–concentration graph seen in Fig. 3.6B. For the majority of drugs eliminated by the liver, this effect is not seen at normal therapeutic doses and only occurs at very high supratherapeutic levels, which is why the kinetics of some drugs in overdose is different from normal. However, one important exception is phenytoin, where saturation of the enzymatic pathway occurs at therapeutic doses. This will be dealt with in the section on phenytoin.

Increasing clearance

The only other situation where first-order elimination is not seen is where clearance increases as the plasma concentration increases (Fig. 3.6C). Under normal circumstances, the plasma protein binding sites available to a drug far outnumber the capacity of the drug to fill those binding sites, and the proportion of the total concentration of drug which is protein bound is constant. However, this situation is not seen in one or two instances (e.g. valproate and disopyramide). For these particular drugs, as the concentration increases the plasma protein binding sites become saturated and the ratio of unbound drug to bound drug increases. The elimination of these drugs increases disproportionate to the total concentration, since elimination is dependent on the unbound concentration.

Clinical applications

Estimation of creatinine clearance

Since many drugs are renally excreted, and the most practical marker of renal function is creatinine clearance, it is often necessary to estimate this in order to undertake dosage adjustment in renal impairment. The usual method is to undertake a 24-h urine collection coupled with a plasma creatinine measurement. The laboratory then estimates the patient’s creatinine clearance. The formula used to determine creatinine clearance is based upon the pharmacokinetic principles in Eq. (3).

The rate of elimination is calculated from the measurement of the total amount of creatinine contained in the 24-h urine sample divided by 24, that is,

image

Using this rate of excretion and substituting the measured plasma creatinine for Css in Eq. (4), the creatinine clearance can be calculated.

However, there are practical difficulties with this method. The whole process is cumbersome and there is an inevitable delay in obtaining a result. The biggest problem is the inaccuracy of the 24-h urine collection.

An alternative approach is to estimate the rate of production of creatinine (i.e. rate in) instead of the rate of elimination (rate out). Clearly this has advantages, since it does not involve 24-h urine collections and requires only a single measure of plasma creatinine. There are data in the literature relating creatinine production to age, weight and sex since the primary source of creatinine is the breakdown of muscle.

Therefore, equations have been produced which are rearrangements of Eq. (4), that is,

image

Rate of production is replaced by a formula which estimates this from physiological parameters of age, weight and sex.

It has been shown that the equation produced by Cockcroft and Gault (1976) appears to be the most satisfactory. A modified version using SI units is shown as

image

where F = 1.04 (females) or 1.23 (males).

There are limitations using only plasma creatinine to estimate renal function. The modification of diet in renal disease (MDRD) formula can be used to estimate glomerular filtration rate (eGFR). This formula uses plasma creatinine, age, sex and ethnicity (Department of Health, 2006).

image

eGFR = glomerular filtration rate (mL/min per 1.73 m2).

The MDRD should be used with care when calculating doses of drugs, as most of the published dosing information is based on Cockcroft and Gault formula. In patients with moderate to severe renal failure, it is best to use the Cockcroft and Gault formula to determine drug dosing.

Digoxin

Distribution

Digoxin is widely distributed and extensively bound in varying degrees to tissues throughout the body. This results in a high apparent volume of distribution. Digoxin volume of distribution can be estimated using the equation 7.3 L/kg (ideal body weight (BWt)) which is derived from population data. However, distribution is altered in patients with renal impairment, and a more accurate estimate in these patients is given by:

image

A two-compartment model best describes digoxin disposition (see Fig. 3.3), with a distribution time of 6–8 h. Clinical effects are seen earlier after intravenous doses, since the myocardium has a high blood perfusion and affinity for digoxin. Sampling for TDM must be done no sooner than 6 h post-dose, otherwise an erroneous result will be obtained.

Theophylline

Theophylline is an alkaloid related to caffeine. It has a variety of clinical effects including mild diuresis, central nervous system stimulation, cerebrovascular vasodilatation, increased cardiac output and bronchodilatation. It is the last which is the major therapeutic effect of theophylline. Theophylline does have some serious toxic effects. However, there is a good plasma concentration–response relationship.

Gentamicin

Practical implications

Since the therapeutic range is based on peak (1 h post-dose to allow for distribution) and trough (pre-dose) concentrations, it is necessary to be able to predict these from any given dosage regimen.

Initial dosage

This may be based on the patient’s physiological parameters. Gentamicin clearance may be determined directly from creatinine clearance. The volume of distribution may be determined from ideal body weight. The elimination constant ke may then be estimated using these parameters in Eq. (6). By substituting ke and the desired peak and trough levels into Eq. (7), the optimum dosage interval can be determined (add on 1 h to this value to account for sampling time). Using this value (or the nearest practical value) and the desired peak or trough value substituted into Eq. (13) or Eq. (14), it is possible to determine the appropriate dose.

Changing dosage

This is not as straightforward as for theophylline or digoxin, since increasing the dose will increase the peak and trough levels proportionately. If this is not desired, then use of pharmacokinetic equations is necessary. By substituting the measured peak and trough levels and the time between them into Eq. (7), it is possible to determine ke (and the half-life from Eq. (8) if required). To estimate the patient’s volume of distribution from actual blood level data, it is necessary to know the Cssmax immediately after the dose (time zero), not the 1 h value which is measured. To obtain this, Eq. (7) may be used, this time substituting the trough level for C2 and solving for C1. Subtracting the trough level from this Cssmax at time zero, the volume of distribution may be determined from Eq. (10). Using these values for ke and Vd, derived from actual blood level data, a new dose and dose interval can be determined as before.

Once daily dosing

There are theoretical arguments for once daily dosing of gentamicin, since aminoglycosides display concentration-dependent bacterial killing, and a high enough concentration to minimum inhibitory concentration (MIC) ratio may not be achieved with multiple dosing. Furthermore, aminoglycosides have a long post-antibiotic effect. Aminoglycosides also accumulate in the kidneys, and once daily dosing could reduce renal tissue accumulation. There have been a number of clinical trials comparing once daily administration of aminoglycosides with conventional administration. A small number of these trials have shown less nephrotoxicity, no difference in ototoxicity, and similar efficacy with once daily administration.

Initial dosage for a once daily regimen is 5–7 mg/kg/day for patients with a creatinine clearance of >60 mL/min. This is subsequently adjusted on the basis of blood levels. However, monitoring of once daily dosing of gentamicin is different to multiple dosing. One approach is to take a blood sample 6–14 h after the first dose and plot the time and result on a standard concentration-time plot (the Hartford nomogram, Nicolau et al., 1995; Fig. 3.7). The position of the individual patient’s point in relation to standard lines on the nomogram indicates what the most appropriate dose interval should be (either 24, 36 or 48 h). Once daily dosing of gentamicin has not been well studied in pregnant or breastfeeding women, patients with major burns, renal failure, endocarditis or cystic fibrosis. Therefore, it cannot be recommended in these groups and multiple daily dosing should be used.

Lithium

Lithium is effective in the treatment of acute mania and in the prophylaxis of manic depression. The mechanism of action is not fully understood, but it is thought that it may substitute for sodium or potassium in the central nervous system. Lithium is toxic, producing dose-dependent and dose-independent side effects. Therefore, TDM is essential in assisting in the management of the dosage.

Phenytoin

Phenytoin is used in the treatment of epilepsy (see Chapter 31). Use is associated with dose-independent side effects which include hirsutism, acne, coarsening of facial features, gingival hyperplasia, hypocalcaemia and folic acid deficiency. However, phenytoin has a narrow therapeutic index and has serious concentration-related side effects.

Practical implications

Since the dose/concentration relationship is non-linear, changes in dose do not result in proportional changes in plasma concentration (see Fig. 3.6B). Using the Michaelis–Menten model, if the plasma concentration is known at one dosage, then Vmax may be assumed to be the population average (7 mg/kg/day), since this is the more predictable parameter, and Km calculated using Eq. (15). The revised values of Km can then be used in Eq. (15) to estimate the new dosage required to produce a desired concentration. Alternatively, a nomogram may be used to assist in dose adjustments (Fig. 3.8).

Care is needed when interpreting TDM data and making dosage adjustments when phenytoin is given concurrently with other anticonvulsants, since these affect distribution and metabolism of phenytoin. Since phenytoin is approximately 90% protein bound, in patients with a low plasma albumin and or uraemia, the free fraction increases and therefore an adjusted total phenytoin should be calculated or a free salivary level taken. To adjust the observed concentration in hypoalbuminaemia the following equation can be applied:

image

Albumin concentration is in g/L.

In ureamic patients with severe renal failure, the unbound fraction is approximately doubled, so the target concentration needs to be half the normal concentration or apply the adjusted concentration equation if albumin level is known.

The oral formulations of phenytoin show good bioavailability. However, tablets and capsules contain the sodium salt (S = 0.9), whereas the suspension and infatabs are phenytoin base (S = 1). Intramuscular phenytoin is slowly and unpredictably absorbed, due to crystallisation in the muscle tissue, and is therefore not recommended. Fosphenytoin, a prodrug of phenytoin, is better absorbed from the intramuscular site. Doses should be expressed as phenytoin equivalent. Fosphenytoin sodium 1.5 mg is equivalent to phenytoin sodium 1 mg.

Carbamazepine

Carbamazepine is indicated for the treatment of partial and secondary generalised tonic-clonic seizures, primary generalised tonic-clonic seizures, trigeminal neuralgia, and prophylaxis of bipolar disorder unresponsive to lithium. There are a number of dose-independent side effects, including various dermatological reactions and, more rarely, aplastic anaemia and Stevens–Johnson syndrome. However, the more common side effects are concentration related.

Phenobarbital

Phenobarbital is indicated in all forms of epilepsy except absence seizures. Although there is a clear concentration–response relationship, routine plasma concentration monitoring is less useful than for other drugs, since tolerance occurs.

Valproate

Sodium valproate as valproic acid in the bloodstream has a broad spectrum of anticonvulsant activity, being useful in generalised absence, generalised tonic–colonic and partial seizures.

Ciclosporin

Ciclosporin is a neutral lipophilic cyclic endecapeptide extracted from the fungus Tolypocladium inflatum gams. It is a potent immunosuppressive agent, used principally to reduce graft rejection after organ and tissue transplantation. The drug has a low therapeutic index, with a number of toxic effects including nephrotoxicity, hepatotoxicity, gastro-intestinal intolerance, hypertrichosis and neurological problems. Efficacy in reducing graft rejection as well as the main toxic effect of nephro- and hepatotoxicity appear to be concentration related.

Practical implications

In addition to the wide inter-patient variability in distribution and elimination pharmacokinetic parameters, absorption of standard formulations of ciclosporin is variable and incomplete (F = 0.2–0.5 in normal subjects). In transplant patients this variation in bioavailability is even greater, and increases during the first few months after transplant. Furthermore, a number of drugs are known to interact with ciclosporin. All these factors suggest that therapeutic drug monitoring will assist in optimum dose selection, but the use of population averages in dose prediction is of little benefit, due to wide inter-patient variation. When using TDM with ciclosporin a number of practical points need to be considered:

Summary pharmacokinetic data for drugs with therapeutic plasma concentrations are listed in Table 3.1

Case studies

Rearranging Eq. (4):

image

References

Cockcroft D.W., Gault M.H. Prediction of creatinine clearance from plasma creatinine. Nephron. 1976;16:31-41.

Crist K.D., Nahata M.C., Ety J. Positive impact of a therapeutic drug monitoring program on total aminoglycoside dose and hospitalisation. Ther. Drug Monit.. 1987;9:306-310.

Department of Health. Estimated Glomerular Filtration Rate (eGFR). London: Department of Health Publications, 2006. Available from: //www.dh.gov.uk/en/Publicationsandstatistics/Publications/PublicationsPolicyAndGuidance/DH_4133020

Evans W.E., Shentag J.J., Jusko W.J., editors. Applied Pharmacokinetics, 3rd edn. Applied Therapeutics. Baltimore: Lippincott Williams & Wilkins. 1992:586-617.

Gjesdal K., Feyzi J., Olssen S.B. Digitalis: a dangerous drug in atrial fibrillation? Analysis of the SPORTIF III and V data. Heart. 2008;94:191-196.

McFadyen M.L., Miller R., Juta M., et al. The relevance of a first world therapeutic drug monitoring service to the treatment of epilepsy in third world conditions. S. Afr. Med. J.. 1990;78:587-590.

Nicolau D.P., Freeman C.D., Belliveau P.P., et al. Experience with a once daily aminoglycoside program administered to 2,184 adult patients. Antimicrob. Agents Chemother.. 1995;39:650-655.

Reid L.D., Horn J.R., McKenna D.A. Therapeutic drug monitoring reduces toxic drug reactions: a meta-analysis. Ther. Drug Monit. 1990;12:72-78.

Touw D.J., Neef C., Thomson A.H., et al. Cost-effectiveness of therapeutic drug monitoring: an update. Eur. J. Hosp. Pharm. Sci.. 2007;13:83-91.