<<

BLUK043-Reid June 29, 2006 17:47

Part 1 Principles of clinical pharmacology

1 BLUK043-Reid June 29, 2006 17:47

2 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

Prior to the twentieth century, medical practice adverse reactions and potentially individualising depended largely on the administration of mix- drug therapy. tures of natural plant or animal substances. These preparations contained a number of pharmacolog- ically active agents in variable amounts. Their ac- Principles of drug action tions and indications were empirical and based (pharmacodynamics) on historical or traditional experience. Their use Pharmacological agents are used in therapeutics to: was rarely based on an understanding of the 1 Cure disease: mechanism of disease or careful measurement of ● Chemotherapy in cancer or leukaemia effect. ● Antibiotics in specific bacterial infections During the last 100 years an increased un- 2 Alleviate symptoms: derstanding has developed of biochemical and ● Antacids in dyspepsia pathophysiological factors that influence disease. ● Non-steroidal anti-inflammatory drugs in The chemical synthesis of agents with well- rheumatoid arthritis characterised, specific actions on cellular mecha- 3 Replace deficiencies: nisms has led to the introduction of many pow- ● Thyroxine in hypothyroidism erful and effective drugs. Additionally, advances ● Insulin in diabetes mellitus in the detection of these compounds in body flu- 4 Prevent or delay end-stage consequences of de- ids have facilitated investigation into the relation- generative diseases, ageing, etc. ships between the dosage regimen, the profile of A drug is a single chemical entity that may be drug concentration against time in body fluids, one of the constituents of a medicine. notably the plasma, and corresponding profiles of A medicine may contain one or more active con- clinical effect. Knowledge of this concentration– stituents (drugs) together with additives to facili- effect relationship and the factors that influence tate administration. drug concentrations are used to determine how much drug an individual patient will require, and how often it should be given. Mechanism of drug action More recently the elucidation of the human Action on a receptor genome with the development of genomics and proteomics has provided new insights and op- A receptor is a specific macromolecule, usually portunities for drug development, understanding a protein, to which a specific group of drugs or

3 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

naturally occurring substances (such as neuro- The biochemical events that result from an transmitters or hormones) can bind. agonist–receptor interaction and which produce An agonist is a substance that stimulates or acti- an effect, are complex. There are many types of vates the receptor to produce an effect. receptors and in several cases subtypes have been identified which are also of therapeutic importance e.g. at the β2-receptor (Table 1.1). An antagonist prevents the action of an agonist but does not have any effect itself. Action on an enzyme e.g. losartan at the angiotensin II receptor Enzymes, like receptors, are protein macro- A partial agonist stimulates the receptor to a lim- molecules with which substrates interact to ited extent, while preventing any further stimula- produce activation or inhibition. Drugs in com- tion by naturally occurring agonists. mon clinical use which exert their effect through

e.g. at the β1-receptor enzyme action generally do so by inhibition.

Table 1.1 Some receptors involved in the action of commonly used drugs.

Receptor Subtype Main actions of natural agonist Drug agonist Drug antagonist

Adrenoceptor α1 Vasoconstriction

α2 Hypotension, sedation Moxonidine

β1 Heart rate Dopamine Metoprolol

β2 Bronchodilation Salbutamol Vasodilation Uterine relaxation Cholinergic Muscarinic Heart rate Atropine Secretion Benzatropine (benztropine) Gut motility Orphenadrine Bronchoconstriction Ipratropium Nicotinic Contraction of striated muscle Suxamethonium Tubocurarine

Histamine H1 Bronchoconstriction Chlorphenamine (chlorpheniramine) Capillary dilation Terfenadine

H2 ↑Gastric acid Cimetidine Ranitidine Famotidine 5-Hydroxy- Fluoxetine Ondansetron tryptamine Fluvoxamine Granisetron Dopamine CNS neurotransmitter Opioid CNS neurotransmitter Morphine, Naloxone pethidine, etc.

4 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

1 Digoxin inhibits the membrane bound Na+/K+ Often the mechanisms have been defined in terms ATPase. of effects on specific receptors or enzymes. In other 2 Aspirin inhibits platelet cyclo-oxygenase. cases chemical action (alkylation) damages DNA or 3 Enalapril inhibits angiotensin-converting en- other macromolecules and results in cell death or zyme. failure of cell division. 4 Selegiline inhibits monoamine oxidase B. 5 Carbidopa inhibits dopa decarboxylase. Dose–response relationship 6 Allopurinol inhibits xanthine oxidase. Drug receptor antagonists and enzyme In clinical practice dose–response relationships inhibitors can act as competitive, reversible rarely follow the classical sigmoid pattern of antagonists or as non-competitive, irreversible experimental studies. It is uncommon for the antagonists. The duration of the effect of drugs upper plateau or maximum effect to be reached of the latter type is much longer than that of the in humans or to be relevant therapeutically. former. Effects of competitive antagonists can be Additionally, variability in the relationship overcome by increasing the dose of endogenous between dose and concentration means that or exogenous agonists, while effects of irreversible it is often difficult to detect a dose–response antagonists cannot usually be overcome. relationship. Consequently, concentration– Atenolol is a competitive β-adrenoceptor antag- response relationships are often more clinically onist used in hypertension and angina. Its effects relevant. last for hours and can be overcome by administer- Dose– (or concentration–) response relation- ing an appropriate dose of a β-receptor agonist like ships may be steep or flat. A steep relationship . implies that small changes in dose will produce Vigabatrin is an irreversible inhibitor of gamma large changes in clinical response or adverse ef- aminobutyric acid (GABA) aminotransferase and is fects, while flat relationships imply that increas- used in epilepsy. Its action and adverse effects may ing the dose will offer little clinical advantage persist for days as a result of irreversible binding to (Fig. 1.1). the target enzyme. The potency of a drug is relatively unimportant; what matters is its efficacy or the maximum ef- fect that can be obtained. In clinical practice the Action on membrane ionic channels maximum therapeutic effect may often be unob- tainable because of the appearance of adverse or The conduction of impulses in nerve tissues and unwanted effects: few, if any, drugs cause a sin- electromechanical coupling in muscle depend on gle pharmacological response. The concentration– the movement of ions, particularly sodium, cal- adverse response relationship is often different in cium and potassium, through membrane chan- shape and position to that of the concentration– nels. Several groups of drugs interfere with these therapeutic response relationship. The difference processes: between the concentration that produces the de- 1 Anti-arrhythmic drugs sired effect and the concentration that causes 2 Calcium slow channel antagonists adverse effects is called the therapeutic index 3 General and local anaesthetics and is a measure of the selectivity of a drug 4 Anticonvulsants. (Fig. 1.2). The shape and position of dose–response curves for a group of patients is variable because of ge- Cytotoxic actions netic, environmental and disease factors. However, Drugs used in cancer or in the treatment of infec- this variability is not solely an expression of differ- tions may kill malignant cells or micro-organisms. ences in response to drugs. It has two important

5 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

100

(a) Effect (%)

(b) Figure 1.1 Schematic examples of a drug (a) with a steep dose– (or concentration–) response relationship in the therapeutic range, e.g. war- Therapeutic range farin as an oral anticoagulant; and (b) 0 a flat dose– (or concentration–) re- sponse relationship within the thera- Dose peutic range, e.g. thiazide diuretics in hypertension.

components: the dose–plasma concentration re- concentration relationships so that this com- lationship and the plasma concentration–effect ponent of the variability in response can be relationship. taken into account when drugs are prescribed for patients with various disease states. For drugs with Dose → Concentration → Effect a narrow therapeutic index it may be necessary With the development of specific and sensi- to measure plasma concentrations to assess the tive chemical assays for drugs in body fluids, it relationship between dose and concentration in has been possible to characterise dose–plasma individual patients.

100 Dose–therapeutic response relationship

Maximum tolerated Dose–adverse adverse effect response relationship

Effect (%) Minimum useful Figure 1.2 Schematic diagram of the effect dose–response relationship for the Therapeutic desired effect (dose–therapeutic re- range sponse) and for an undesired adverse 0 effect. The therapeutic index is the Dose extent of displacement of the two curves within the normal dose range.

6 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

Table 1.2 Several drugs that undergo extensive Clinical pharmacology: What are kinetics first-pass metabolism. and dynamics?

The description of a drug concentration profile against Analgesics Drugs acting on CNS time is known as pharmacokinetics and its application in Aspirin Clomethiazole clinical practice is clinical pharmacokinetics (Chapter 2). (chlormethiazole) The residual variability in the relationship between dose Morphine Chlorpromazine and response is the concentration–effect component—a Paracetamol true expression of drug response, and a measure of the Pethidine Levodopa sensitivity of a patient to a drug. This is known as phar- macodynamics. Clinical pharmacology seeks to explore Cardiovascular drugs the factors that underlie variability in pharmacokinetics Glyceryl trinitrate Respiratory drugs and pharmacodynamics and to use this information to optimise drug therapy for individual patients. Isoprenaline Salbutamol Isosorbide dinitrate Terbutaline Lidocaine (lignocaine) Oral contraceptives Metoprolol Principles of pharmacokinetics Nifedipine Prazosin Absorption Drug absorption after oral administration has two Verapamil major components: absorption rate and bioavail- ability. Absorption rate is controlled partially by the physicochemical characteristics of the drug but of gastrointestinal epithelium that is drained by in many cases is modified by the formulation. A re- veins forming part of the hepatoportal system. duction in absorption rate can lead to a smoother Consequently, even if they are well absorbed, drugs concentration–time profile with a lower potential must pass through the liver before reaching the for concentration-dependent adverse effects and systemic circulation. For drugs that are suscepti- may allow less frequent dosing. ble to extensive hepatic metabolism, a substantial Bioavailability is the term used to describe the proportion of an orally administered dose can be fraction of the dose that is absorbed into the sys- metabolised before it ever reaches its site of phar- temic circulation and is usually designated F . It can macological action. Drugs with a high first-pass range from 0 to 1 (0–100%) and depends on a num- metabolism are listed in Table 1.2. ber of physicochemical and clinical factors. Low The importance of first-pass metabolism is two- bioavailability may occur if the drug has low sol- fold: ubility or is destroyed by the acid in the stomach. 1 It is one of the reasons for apparent differ- Changing the formulation can affect the bioavail- ences in drug absorption between individuals. ability of a drug and it can also be altered by Even healthy people show considerable variation food or the co-administration of other drugs. For in liver metabolising capacity. example, antacids can reduce the absorption of 2 In patients with severe liver disease first-pass quinolone antibiotics by binding them in the gut. metabolism may be dramatically reduced, leading Other factors influencing bioavailability include to the appearance of greater amounts of parent metabolism by gut flora, the intestinal wall or the drug in the systemic circulation. liver. First-pass metabolism refers to metabolism of a Distribution drug that occurs en route from the gut lumen to the systemic circulation. For the majority of drugs Once a drug has gained access to the bloodstream given orally, absorption occurs across the portion it begins to distribute to the tissues. The extent

7 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

of this distribution depends on a number of fac- Plasma protein binding tors including plasma protein binding, the pKa In the blood, a proportion of a drug is bound to of the drug, its partition coefficient in fatty tis- plasma proteins—mainly albumin (acidic drugs) sue and regional blood flow. The volume of dis- and α1-acid glycoprotein (basic drugs). Only the tribution VD is the apparent volume of fluid into unbound, or free, fraction distributes because which a drug distributes based on the amount of the protein-bound complex is too large to pass drug in the body and the measured concentration through membranes. Movement of the drug be- in the plasma or serum. If a drug was wholly con- tween the blood and other tissues proceeds until fined to the plasma, VD would equal the plasma equilibrium is established between the unbound volume—approximately 3 l in an adult. If, on the drug in plasma and the drug in tissues. It is other hand, the drug was distributed throughout the unbound portion that is generally responsi- the body water, VD would be approximately 42 l. ble for clinical effects—both the target response In reality, drugs are rarely distributed into phys- and the unwanted adverse effects. Changes in pro- iologically relevant volumes. If most of the drug tein binding (e.g. resulting from displacement in- is bound to tissues, the plasma concentration will teractions) generally lead to a transient increase in be low and the apparent VD will be high, while free concentration and are rarely clinically relevant high plasma protein binding will tend to main- because the equilibrium becomes re-established tain high concentrations in the blood and a low with the same unbound concentration. However, VD will result. For the majority of drugs, VD de- a lower total concentration will be present and pends on the balance between plasma binding and the measurement might be misinterpreted if the sequestration or binding by various body tissues, higher free fraction is not taken into account. This for example, muscle and fat. Volume of distribu- is a common problem with the interpretation of tion can vary therefore from relatively small val- phenytoin concentrations, where free fraction can ues (e.g. an average of 0.14 l/kg body weight for range from 10% in a normal patient to 40% in a aspirin) to large values (e.g. an average of 200 l/kg patient with hypoalbuminaemia and renal impair- body weight for chloroquine) (Table 1.3). ment.

Table 1.3 Average volumes of distribution of some commonly used drugs. Clinical relevance of volume of distribution Drug Volume of distribution (l/kg) Knowledge of volume of distribution (VD) can be Chloroquine 200 used to determine the size of a loading dose if an Nortriptyline 20 immediate response to treatment is required. This Digoxin 7 Propranolol 4 assumes that therapeutic success is closely related Phenytoin 0.65 to the plasma concentration and that there are no Theophylline 0.50 adverse effects if a relatively large dose is suddenly Gentamicin 0.25 administered. It is sometimes employed when drug Aspirin 0.14 response would take many hours or days to de- Warfarin 0.10 velop if the regular maintenance dose was given from the outset, e.g. digoxin. In general, a small VD occurs when: 1 Lipid solubility is low. A loading dose can be calculated as follows: 2 There is a high degree of plasma protein binding. Loading dose = V × Desired concentration 3 There is a low level of tissue binding. (Eqn. 1.1) A high VD occurs when: 1 Lipid solubility is high. In practice, because most values for VD are re- 2 There is a low degree of plasma protein binding. lated to weight, this calculation is often simplified 3 There is a high level of tissue binding. to a mg/kg dose.

8 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

Clearance Equations 1.2 and 1.3 highlight the important fact that if an estimate of clearance is available, it Clearance is the sum of all drug-eliminating pro- can be used to determine the maintenance dose for cesses, principally determined by hepatic meta- any desired Css, thus bolism and renal excretion. It can be defined as

the theoretical volume of fluid from which a drug Infusion rate = Clearance × Desired Cssaverage is completely removed in a given period of time. (Eqn. 1.4a) When a drug is administered continuously by intravenous infusion or repetitively by mouth, a or for oral therapy, balance is eventually achieved between its input (dosing rate) and its output (the amount elimi- maintenance dose = Clearance nated over a given period of time). This balance × Desired Css gives rise to a constant amount of drug in the body average which depends on the dosing rate and clearance. × Dosage interval/F This amount is reflected in the plasma or serum as (Eqn. 1.4b) a steady-state concentration (Css). A constant rate intravenous infusion will clearly yield a constant Clearance depends critically on the efficiency Css, while a drug administered orally at regular in- with which the liver and/or kidneys can eliminate tervals will result in fluctuation between peak and a drug; it will vary in disease states that affect these trough concentrations (Fig. 1.3). organs per se, or that affect the blood flow to these The average Css in any dosage interval may be organs. In stable clinical conditions, clearance re- approximated by the concentration one-third of the mains constant and Eqns. 1.2 and 1.3 show that way between the trough and the peak. the Cssaverage is directly proportional to dose rate. The relationship between the average Css, drug The important implication is that if the dose rate input and drug output for a constant rate infusion is doubled, the Cssaverage doubles: if the dose rate is can be written as halved, the Cssaverage is halved. This is illustrated Input rate Infusion rate in Fig. 1.4. If each Cssaverage is plotted against its Cssaverage = = Output rate Clearance corresponding dose rate, the direct proportional- (Eqn. 1.2) ity becomes obvious (Fig. 1.5). In pharmacokinetic or for oral therapy, terms this is referred to as a first-order or linear F × Dose process, and results from the fact that the rate of Cssaverage = Clearance × Dosage interval elimination is proportional to the amount of drug (Eqn. 1.3) present in the body.

25

20

15

10 Concentration 5

Figure 1.3 Steady-state concen- 0 tration–time profile for an oral 0 5 101520253035404550 dose (——) and a constant rate Time (h) intravenous infusion (- - - - -).

9 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

Css 3 (infusion 3) 90

Css 2 (infusion 2) 60

Css 1 (infusion 1) 30 Concentration (mg/l) Figure 1.4 Plots of concentration vs. time for three infusions allowed to reach steady state. Infusion 2 is at a rate twice that of infusion 1; infu- 0 sion 3 is at a rate three times that 0122436 48 60 of infusion 1. The three steady-state concentrations (C ss 1, 2 and 3) are di- Time (h) rectly proportional to the correspond- ing infusion rates.

Single intravenous bolus dose the drug distributes instantaneously into its vol- ume of distribution VD, then its initial concentra- A number of other important pharmacokinetic tion C0 depends only on the dose (D) and VD; thus principles can be appreciated by considering the concentrations that result following a single intra- D C0 = (Eqn. 1.5) venous bolus dose (see Fig. 1.6a). If we assume that VD

90

60

30 Steady-state concentration (mg/l)

0 Figure 1.5 Three steady-state con- 0 30 60 90 centrations plotted against corre- (infusion 1) (infusion 2) (infusion 3) sponding infusion rates showing the Infusion rate (mg/h) linear relationship between dose and C ssaverage.

10 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

100

90 Input Co = D/V (drug dosage) 80

70

60

50 V

40 Concentration 30 Output 20 (drug elimination) 10

0 01234 56 798 (a) Time (half-lives) Figure 1.7 The body depicted as a single compartment of volume V .

100 This is based on the concept that the body can be depicted as a single homogeneous compartment Slope = −k of volume V, as shown in Fig. 1.7. The concentra- tion will then decline by a constant proportion per 10 unit time, giving rise to an exponential decline. The concentration at any time t after the dose can therefore be determined from the exponential ex- pression Concentration 1 D − C(t) = e kt (Eqn. 1.6) V where k is the elimination rate constant of the drug, t is any time after drug administration and −kt 0.1 e is the fraction of drug remaining at time t. 01234 56 798 If the concentrations are plotted on a logarithmic (b) Time (half-lives) scale, a linear decline will be obtained with slope-k and intercept ln D/V; thus Figure 1.6 (a) Plot of concentration vs. time after a D bolus intravenous injection. The intercept on the y- In C(t) = In − kt (Eqn. 1.7) V (concentration) axis, C , is the concentration resulting 0 / from the instantaneous injection of the bolus dose. Semi-logarithmic graph paper allows C0(D V) (b) Semi-logarithmic plot of concentration vs. time after a to be determined directly (Fig. 1.6b). k represents bolus intravenous injection. The slope of this line is −k; the the constant fraction of the volume of distribution elimination rate constant (Eqns. 1.6 and 1.7) and the elim- from which a drug is eliminated in a given period ination half-life of the drug can be easily determined from of time and therefore depends on both clearance such a plot by noting the time at which the concentration has fallen to half its original value. and volume of distribution; thus Clearance k = (Eqn. 1.8) Volume of distribution k can also be expressed in terms of the half-life of a

11 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

toxic) peaks or low, ineffective troughs. Excessive 4 fluctuations in the concentration–time profile can be prevented by giving the drug at intervals of less 3 than one half-life or by using a slow-release formu- lation. 2

Concentration 1 Linear vs. non-linear kinetics

In the discussion on clearance, it was pointed out 0 010203040 50 6070 that the hallmark of linear pharmacokinetics is the Time (h) proportionality between dose rate and steady-state concentration. This arises because the rate of elimi- Figure 1.8 Plot of concentration vs. time illustrating the nation is proportional to the amount of drug in the accumulation to steady state when a drug is administered body, while the clearance remains constant. This by regular oral doses. is not, however, always the case as is exemplified by the anticonvulsant drug phenytoin. When the enzymes responsible for metabolism reach a point drug. The half-life t1/2 is the time required for the of saturation, the rate of elimination, in terms of plasma concentration to fall to half of its original amount of drug eliminated in a given period of value and can be derived either graphically (Fig. time, does not increase in response to an increase 1.6) or from the expression in concentration (or an increase in the amount of In2 drug in the body) but becomes constant. This gives t1/2 = (Eqn. 1.9) k rise to non-linear or zero-order kinetics. where ln 2 is the natural logarithm of 2, or 0.693. The general relationship between drug concen- It can be used to predict the time at which steady tration (C ) and rate of metabolism is shown in Fig. state will be achieved after starting a regular treat- 1.9. The maximum rate at which the enzymes can ment schedule or after any change in dose. As a function, Vmax, corresponds to the plateau attained rule, in the absence of a loading dose, steady state by the curve. is attained after four to five half-lives (Fig. 1.8). The equation relating the rate of metabolism to Furthermore, when toxic drug levels have been in- C is the Michaelis–Menten equation advertently produced, it is very useful to estimate V × C how long it will take for such levels to reach the Rate of metabolism = max (Eqn. 1.10) K × C therapeutic range, or how long it will take for all m the drug to be eliminated once the drug has been and the fundamental difference between linear stopped. Usually, elimination is effectively com- and non-linear kinetics can be appreciated by con- plete after four to five half-lives (Fig. 1.6). sidering two extreme cases. The elimination half-life can also be used to 1 The serum concentration is considerably less

determine dosage intervals to achieve a target than Km. In this case, the Michaelis–Menten equa- concentration–time profile. For example, in order tion can be approximated to to obtain a gentamicin peak of 8 mg/l and a trough V × C of 0.5 mg/l in a patient with an elimination half- Rate of metabolism = max (Eqn. 1.11) K life of 3 h, the dosage interval should be 12 h. (The m

concentration will fall from 8 mg/l to 4 mg/l in 3 where Vmax/Km is a constant. This means that the h, to 2 mg/l in 6 h, to 1 mg/l in 9 h and to 0.5 mg/l rate of change of concentration is then propor- in 12 h.) However, for many drugs, dosage regi- tional to the concentration (linear kinetics). mens should be designed to maintain concentra- 2 The serum concentration is considerably greater

tions within a range that avoids high (potentially than Km.

12 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

Vmax Figure 1.9 Diagrammatic represen- tation of the general relationship be- tween drug concentration C and the C rate of metabolism. Vmax is the max- d /dt imum velocity at which the drug- metabolising enzyme can function and is a constant (with units of 50% Vmax mass/time). K m is the concentration at which Vmax is 50%. The K m is usually much higher than therapeu- tic concentrations and the rate of metabolism vs. C is essentially linear (Eqn. 1.11). With a few drugs, no- tably phenytoin, therapeutic plasma concentrations are in the region of 0 K so that rate of metabolism vs. C m K Concentration (mg/l) is non-linear and governed by the re- m lationship shown in Eqn. 1.10.

In this case, the Michaelis–Menten equation can Comment. The clinical relevance of non-linear ki- be approximated to netics is that a small increase in dose can lead to a large increase in concentration. This is particu- = Rate of metabolism Vmax (Eqn. 1.12) larly important when toxic side effects are closely related to concentration, as with phenytoin. which indicates that the rate of elimination is a constant. At steady state, dose rate can be substituted for Principles of drug elimination rate of metabolism, i.e. Drug metabolism × = Vmax Css Steady-state dose rate Drugs are eliminated from the body by two princi- Km + Css (Eqn. 1.13) pal mechanisms: (i) liver metabolism and (ii) renal excretion. Drugs that are already water-soluble are

For phenytoin, Vmax has a typical value of 7.2 generally excreted unchanged by the kidney. Lipid- mg/kg per day and Km has a typical value of soluble drugs are not easily excreted by the kidney 4.4 mg/l (17.6 μmol/l). In the case of pheny- because, following glomerular filtration, they are toin, the range of concentrations used clinically largely reabsorbed from the proximal tubule. The

encompasses and exceeds Km. Consequently, the first step in the elimination of such lipid-soluble relationship between the steady-state concentra- drugs is metabolism to more polar (water-soluble) tion and dose rate will alter as the concentra- compounds. This is achieved mainly in the liver, tion changes. At low concentrations, the increase but can also occur in the gut and may contribute in concentration will be proportional to the dose to first-pass elimination. Metabolism generally oc- rate (linear pharmacokinetics). At higher concen- curs in two phases: trations, the increase will be much greater than Phase 1 Mainly oxidation (sometimes reduction or would have been anticipated (non-linear pharma- hydrolysis) to a more polar compound. cokinetics). Steady state will not be achieved if Phase 2 Conjugation, usually with glucuronic acid

the dose rate exceeds Vmax. This can be seen in or sulphate, to make the compound substan- Fig. 1.10. tially more polar.

13 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

80

60

40

20 Steady-state concentration (mg/l)

0 Figure 1.10 0 100 200 300 400 500 The C ss vs. dose rate re- lationship for phenytoin. This is gov- Dose rate (mg/day) erned by Michaelis–Menten kinetics (Eqn. 1.13).

Phase 1 metabolism Phase 2 reactions

Oxidation can occur in various ways, including These involve the addition of small endogenous aromatic or aliphatic hydroxylation, oxygenation molecules to the parent drug, or to its phase 1 at carbon, nitrogen or sulphur atoms and N- and O- metabolite, and almost always lead to abolition of dealkylation. These reactions are catalysed by the pharmacological activity. Multiple forms of conju- cytochrome P-450-dependent system of the endo- gating enzymes are also known to exist, although plasmic reticulum. Knowledge of P-450, which ex- these have not been investigated to the same ex- ists as a superfamily of similar enzymes (isoforms), tent as the P-450 system. has increased greatly recently. The P-450 superfam- ily is divided into a number of families and subfam- Metabolic drug interactions ilies, where genes encoding for proteins within a family have at least 40% nucleotide sequence ho- The wide range of drugs metabolised by the P-450 mology and subfamilies have over 65% homology. system provides the opportunity for interactions Although numerous P-450 isoforms are present in of two types, namely enzyme induction and inhi- human tissue, only a few of these have a major bition. role in the metabolism of drugs. These enzymes, which display a distinct but overlapping substrate Induction specificity, are listed in Table 1.4. Enzyme induction, which may be defined as the in- Phase 1 metabolites usually have only minor crease in amount and activity of drug-metabolising structural differences from the parent drug, but enzymes, is a consequence of new protein synthe- may exhibit totally different pharmacological ac- sis resulting from prolonged exposure to the in- tions. For example, the metabolism of azathio- ducing drug. While a drug may induce its own prine produces the powerful antimetabolite 6- metabolism, it can also accelerate the metabolism mercaptopurine. and clearance of unrelated compounds. Many

14 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

Table 1.4 Major human P-450 enzymes involved in drug metabolism.

Major human P-450s Typical substrates

CYP1A2 Theophylline, caffeine, tacrine, fluvoxamine, oestradiol, phenacetin (R)-warfarin CYP2C9 (S)-Warfarin, tolbutamide, glipizide, losartan, ibuprofen, diclofenac, phenytoin CYP2C19 (S)-Mephenytoin, omeprazole, diazepam, citalopram, proguanil, moclobemide CYP2D6 (S)-Metoprolol, bufuralol, dextromethorphan, fluoxetine, , nortryptiline CYP2E1 Enflurane, halothane, chlorzoxazone, ethanol CYP3A4 Astemizole, terfenadine, cisapride, pimozide, nisoldipine, midazolam, indinavir, lovastatin, St. John’s wort

compounds are known to act as enzyme inducers Inhibition in animals at toxicological dose levels, but rela- Concurrently administered drugs can also lead tively few drugs produce clinically significant in- to inhibition of enzyme activity, with many P- duction in humans when used at therapeutic dose 450 inhibitors showing considerable isoform se- levels. lectivity. Some of the most clinically relevant in- The compounds shown in Table 1.5 are the hibitors are listed in Table 1.6, together with the most potent enzyme inducers in clinical use and isoform inhibited. For example, ketoconazole de- have produced numerous clinically significant creases the metabolism of the CYP3A4 substrate, drug interactions, related primarily to increases terfenadine, leading to potentially dangerous ad- in the metabolism of CYP2C9, CYP2C19 and verse effects, e.g. QT interval prolongation and tor- CYP3A4 substrates. For example, the anticonvul- sades de pointes. sants phenytoin and carbamazepine, as well as the herbal remedy St. John’s wort, induce the enzymes that metabolise the constituents of oral contracep- Table 1.6 P-450 inhibitors involved in drug interactions. tives. If a woman receiving an oral contraceptive starts taking one of these drugs, the metabolism Major human P-450s Typical inhibitors of the oestrogen and progestogen in the oral con- CYP1A2 Furafylline, fluvoxamine, traceptive increases, with the risk of contraceptive ciprofloxacin failure. Enzyme induction is not, however, limited CYP2C9 Fluconazole, to drug administration. Cigarette smoking, for ex- ketoconazole, ample, results in enzyme induction with increased sulfaphenazole metabolism of CYP1A2 substrates, such as theo- CYP2C19 Omeprazole, phylline, and ethanol is an inducer of CYP2E1. ketoconazole, cimetidine CYP2D6 , fluoxetine, ritonavir Table 1.5 Some of the most potent enzyme inducers in CYP2E1 Disulfiram humans. CYP3A4 Ketoconazole, Carbamazepine itraconazole, ritonavir, Phenytoin erythromycin, Rifampicin diltiazem

15 BLUK043-Reid June 29, 2006 17:47

Chapter 1 Pharmacodynamics and pharmacokinetics

Table 1.7 Major enzymes displaying genetic polymorphism.

Enzyme Typical substrates Characteristics

CYP2C19 (S)-Mephenytoin, diazepam, About 2–5% of white people are poor omeprazole metabolisers, but 18–23% of Japanese people have this phenotype CYP2D6 Propafenone, flecainamide, About 7% of white people are poor metabolisers, desipramine but this frequency is only about 2% in black Americans and <1% in Japanese/Chinese N-Acetyl- transferase Hydralazine, sulphonamides, About 50% of white people are slow acetylators isoniazid, procainamide

As with induction, P-450 inhibition is not lim- consequence of environmental factors, including ited to drug administration. Grapefruit juice is the influence of inducers and inhibitors, the main a fairly potent inhibitor of CYP3A4 activity and factor contributing to interindividual variability produces clinically significant interactions with a in metabolism is the underlying genetic basis of number of drugs, including midazolam, simvas- the drug-metabolising enzymes. Although there is tatin and terfenadine. This type of information, probably a genetic component in the control of together with some knowledge of the enzymes in- most P-450 enzymes, some enzymes (e.g. CYP2C19 volved in a particular drug’s clearance, makes it and CYP2D6) actually show genetic polymor- much easier to understand and predict drug inter- phism. This results in distinct subpopulations of actions. poor and extensive metabolisers, where the poor Comment. Enzyme induction produces clinical metabolisers are deficient in that particular en- changes over days or weeks, but the effects of en- zyme. There are a number of enzymes under poly- zyme inhibition are usually observed immediately. morphic control and some clinically important In most circumstances, these changes are mani- examples are shown in Table 1.7. As with enzyme fest as decreases in efficacy resulting from induc- inhibition, genetic polymorphism is primarily a tion, or as increases in adverse effects resulting concern for drugs that have a narrow therapeutic from inhibition. Clinical relevance occurs when index and that are metabolised largely by a sin- drug therapy needs to be altered to avoid the gle polymorphic enzyme. In such cases, the phe- consequences of the drug interaction and this is notype of the patient should be determined and most common and most serious in compounds lower doses of the drug used, or alternative ther- that have a narrow therapeutic index. Clearly, pro- apy should be considered. nounced enzyme inhibition, which may result in plasma concentrations of the inhibited drug be- ing many times higher than intended, can be a Renal excretion major safety issue. For example, co-administration Three processes are implicated in renal excretion of ketoconazole or ritonavir with the hypnotic of drugs: drug midazolam increases the midazolam plasma 1 Glomerular filtration. This is the most common AUC by 15–20 times, a situation which should be route of renal elimination. The free drug is cleared avoided. by filtration and the protein-bound drug remains in the circulation where some of it dissociates to restore equilibrium. Genetic factors in metabolism 2 Active secretion in the proximal tubule. Both The rate at which healthy people metabolise drugs weak acids and weak bases have specific secre- is variable. Although part of this variability is a tory sites in proximal tubular cells. Penicillins are

16 BLUK043-Reid June 29, 2006 17:47

Pharmacodynamics and pharmacokinetics Chapter 1

eliminated by this route, as is about 60% of pro- and bases are reabsorbed, which in turn determines cainamide. the degree of ionisation. 3 Passive reabsorption in the distal tubule. This oc- If renal function is impaired, for example by dis- curs only with un-ionised, i.e. lipid-soluble, drugs. ease or old age, then the clearance of drugs that Urine pH determines whether or not weak acids normally undergo renal excretion is decreased.

17