General Principles of Pharmacology

Published on 27/02/2015 by admin

Filed under Anesthesiology

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 1986 times

General Principles of Pharmacology

HOW DO DRUGS ACT?

Drugs produce their effects on biological systems by several mechanisms; these include physicochemical action, activity at receptors and inhibition of reactions mediated by enzymes.

Physicochemical Properties

Sodium citrate is an alkali and neutralizes acid; it is often administered orally to reduce the likelihood of pneumonitis after regurgitation of gastric contents. Chelating agents (chel is the Greek word for a crab’s claw) combine chemically with metal ions, reducing their toxicity and enhancing elimination, usually in the urine. Such drugs include desferrioxamine (chelates iron and aluminium), dicobalt edetate (cyanide toxicity), sodium calcium edetate (lead) and penicillamine (copper and lead). Stored blood contains a citrate-based anticoagulant which prevents clotting; this chelates calcium ions and may cause hypocalcaemia after massive blood transfusion. Phenol and alcohol denature proteins; they are used occasionally to produce prolonged or permanent nerve blockade.

Action on Receptors

A receptor is a complex structure on the cell membrane which can bind selectively with endogenous compounds or drugs, resulting in changes within the cell which modify its function. These include changes in selective ion channel permeability (e.g. acetylcholine, glutamate, GABA receptors), cyclic adenosine monophosphate (e.g. opioid, β, α2 and dopamine receptors), cyclic guanosine monophosphate (e.g. atrial natriuretic peptide receptor), inositol phosphate and diacylglycerol (e.g. α1, angiotensin AT1, endothelin, histamine H1 and vasopressin V1 receptors) and nitric oxide (e.g. muscarinic M3 receptor).

A compound which binds to a receptor and changes intracellular function is termed an agonist. The classic dose–response relationship of an agonist is shown in Figure 1.1. As the concentration of the agonist increases, a maximum effect is reached as the receptors in the system become saturated (Fig. 1.1A). Conventionally, log dose is plotted against effect, resulting in a sigmoid curve which is approximately linear between 20 and 80% of maximum effect (Fig. 1.1B). Three agonists are shown in Figure 1.2. Agonist A produces 100% effect at a lower concentration than agonist B. Therefore, compared with A, agonist B is less potent but has similar efficacy. Drug C is termed a partial agonist as the maximum effect is less than that of A or B. Buprenorphine is a partial agonist (at the μ-opioid receptor), as are some of the β-blockers with intrinsic activity, e.g. oxprenolol, pindolol, acebutalol, celiprolol.

Antagonists combine selectively with the receptor but produce no effect. They may interact with the receptor in a competitive (reversible) or non-competitive (irreversible) fashion. In the presence of a competitive antagonist, the dose–response curve of an agonist is shifted to the right but the maximum effect remains unaltered (Fig. 1.3A). Examples of this effect include the displacement of morphine by naloxone and endogenous catecholamines by β-blockers.

A non-competitive (irreversible) antagonist also shifts the dose–response curve to the right but, with increasing concentrations, reduces the maximum effect (Fig. 1.3B). For example, the α1-antagonist phenoxybenzamine, used in the preoperative preparation of patients with phaeochromocytoma, has a long duration of action because of the formation of stable chemical bonds between drug and receptor.

The relationship between drug dose and response is often described by a Hill plot (Fig. 1.4). A typical agonist such as that shown in Figure 1.1 produces a straight line with a slope (i.e. Hill coefficient) of + 1.

Action on Enzymes

Drugs may act by inhibiting the action of an enzyme or competing for its endogenous substrate. Reversible inhibition is the mechanism of action of edrophonium (acetylcholinesterase), aminophylline (phosphodiesterase) and captopril (angiotensin-converting enzyme). Irreversible enzyme inhibition occurs when a stable chemical bond is formed between drug and enzyme, resulting in prolonged or permanent inactivity e.g. omeprazole (gastric hydrogen-potassium ATPase), aspirin (cyclo-oxygenase) and organophosphorus compounds (acetylcholinesterase).

However, the interaction between drug and enzyme may be more complex than this simple classification implies. For example, neostigmine inhibits acetylcholinesterase in a reversible manner, but the mechanism of action is more akin to that of an irreversible drug because neostigmine forms covalent chemical bonds with the enzyme.

THE BLOOD–BRAIN BARRIER AND PLACENTA

Many drugs used in anaesthetic practice must cross the blood–brain barrier in order to reach their site of action. The brain is protected from most potentially toxic agents by tightly overlapping endothelial cells which surround the capillaries and interfere with passive diffusion. In addition, enzyme systems are present in the endothelium which break down many potential toxins. Consequently, only relatively small, highly lipid-soluble molecules (e.g. intravenous and volatile anaesthetic agents, opioids, local anaesthetics) have access to the central nervous system (CNS). Compared with most opioids, morphine takes some time to reach its site of action because it has a relatively low lipid solubility. Highly ionized drugs (e.g. muscle relaxants, glycopyrronium) do not cross the blood–brain barrier.

The chemoreceptor trigger zone is situated in the area postrema near the base of the fourth ventricle (see Ch 42). It is not protected by the blood–brain barrier because the capillary endothelial cells are not bound tightly in this area and allow relatively free passage of large molecules. This is an important afferent limb of the vomiting reflex and stimulation of this area by toxins or drugs in the blood or cerebrospinal fluid often leads to vomiting. Many antiemetics act at this site.

The transfer of drugs across the placenta is of considerable importance in obstetric anaesthesia (see Ch 35). In general, all drugs which affect the CNS cross the placenta and affect the fetus. Highly ionized drugs (e.g. muscle relaxants) pass across less readily.

PLASMA PROTEIN BINDING

Many drugs are bound to proteins in the plasma. This is important because only the unbound portion of the drug is available for diffusion to its site of action. Changes in protein binding may have significant effects on the active unbound concentration of a drug, and therefore its actions.

Albumin is the most important protein in this regard and is responsible mainly for the binding of acidic and neutral drugs. Globulins, especially α1-glycoprotein, bind mainly basic drugs. If a drug is highly protein bound (> 80%), any change in plasma protein concentration or displacement of the drug by another with similar binding properties may have clinically significant effects. For example, most NSAIDs displace warfarin, phenytoin and lithium from plasma binding sites, leading to potential toxicity.

Plasma albumin concentration is often decreased in the elderly, in neonates and in the presence of malnutrition, liver, renal or cardiac failure and malignancy. α1-Glycoprotein concentration is decreased during pregnancy and in the neonate but may be increased in the postoperative period and other conditions such as infection, trauma, burns and malignancy.

METABOLISM

Most drugs are lipid-soluble and many are metabolized in the liver into more ionized compounds which are inactive pharmacologically and excreted by the kidneys. However, metabolites may be active (Table 1.1). The liver is not the only site of metabolism. For example, succinylcholine and mivacurium are metabolized by plasma cholinesterase, esmolol by erythrocyte esterases, remifentanil by tissue esterases and, in part, dopamine by the kidney and prilocaine by the lungs.

TABLE 1.1

Examples of Active Metabolites

Drug Metabolite Action
Morphine Morphine-6-glucuronide Potent opioid agonist
Diamorphine 6-Monoacetylmorphine Morphine Opioid agonist
Meperidine (pethidine) Normeperidine (norpethidine) Epileptogenic
Codeine Morphine Opioid agonist
Diazepam Desmethyldiazepam
Temazepam
Oxazepam
Sedative
Tramadol O-desmethyltramadol Opioid agonist
Parecoxib Valdecoxib COX-2 specific inhibitor

A substance is termed a prodrug if it is inactive in the form in which it is administered, pharmacological effects being dependent on the formation of active metabolites. Examples of this are codeine (morphine), diamorphine (6-monoacetylmorphine, morphine), chloral hydrate (trichlorethanol) and parecoxib (valdecoxib). Midazolam is ionized and dissolved in an acidic solution in the ampoule; after intravenous injection and exposure in the blood to pH 7.4, the molecule becomes lipid-soluble.

Drugs undergo two types of reactions during metabolism: phase I and phase II. Phase I reactions include reduction, oxidation and hydrolysis. Drug oxidation occurs in the smooth endoplasmic reticulum, primarily by the cytochrome P450 enzyme system. This system and other enzymes also perform reduction reactions. Hydrolysis is a common phase I reaction in the metabolism of drugs with ester groups (e.g. remifentanil, succinylcholine, atracurium, mivacurium). Amide drugs often undergo hydrolysis and oxidative N-dealkylation (e.g. lidocaine, bupivacaine).

Phase II reactions involve conjugation of a metabolite or the drug itself with an endogenous substrate. Conjugation with glucuronic acid is a major metabolic pathway, but others include acetylation, methylation and conjugation with sulphate or glycine.

Enzyme Induction and Inhibition

Some drugs may enhance the activity of enzymes responsible for drug metabolism, particularly the cytochrome P450 enzymes and glucuronyl transferase. Such drugs include phenytoin, carbamazepine, phenylbutazone, barbiturates, ethanol, steroids and some inhalational anaesthetic agents (halothane, enflurane). Cigarette smoking also induces cytochrome P450 enzymes.

Drugs with mechanisms of action other than on enzymes may also interfere significantly with enzyme systems. For example, etomidate inhibits the synthesis of cortisol and aldosterone – an effect which may explain the increased mortality in critically ill patients which occurred when it was used as a sedative in intensive care. Cimetidine is a potent enzyme inhibitor and may prolong the elimination of drugs such as diazepam, propranolol, oral anticoagulants, phenytoin and lidocaine. Troublesome interactions with enzyme systems are less of a problem with new drugs; if significant enzyme interaction is discovered in the early stages of development, the drug is usually abandoned.

DRUG EXCRETION

Ionized compounds with a low molecular weight (MW) are excreted mainly by the kidneys. Most drugs and metabolites diffuse passively into the proximal renal tubules by the process of glomerular filtration, but some are secreted actively (e.g. penicillins, aspirin, many diuretics, morphine, lidocaine and glucuronides). Ionization is a significant barrier to reabsorption at the distal tubule. Consequently, basic drugs or metabolites are excreted more efficiently in acid urine and acidic compounds in alkaline urine.

Some drugs and metabolites, particularly those with larger molecules (MW > 400 D), are excreted in the bile (e.g. glycopyrronium, vecuronium, pancuronium and the metabolites of morphine and buprenorphine). Ventilation is responsible for excretion of volatile anaesthetic agents.

PHARMACOKINETIC PRINCIPLES

Pharmacokinetics is the study of what happens to drugs after they have been administered. In contrast, pharmacodynamics is concerned with their effects on biological systems. An understanding of the basic principles of pharmacokinetics is an important aid to the safe use of drugs in anaesthesia, pain management and intensive care medicine. Pharmacokinetics is an attempt to fit observed changes in plasma concentration of drugs into mathematical equations which may then be used to predict concentrations under various circumstances.

Derived values describing volume of distribution (V), clearance (Cl) and half-life (t1/2) give an indication of the likely properties of a drug. However, even in healthy individuals of the same sex, weight and age, there is significant variability which makes precise prediction very difficult. It is important to remember that the accepted pharmacokinetic values of drugs are usually the mean of a wide range of observations.

Volume of Distribution

Volume of distribution is a good example of the abstract nature of pharmacokinetics; it is not a real volume but merely a concept which helps us to understand what we observe. Nevertheless, it is a very useful notion which enables us to predict certain properties of a drug and also calculate other pharmacokinetic values.

Imagine that a patient receiving an intravenous dose of an anaesthetic induction agent is a bucket of water and that the drug is distributed evenly throughout the water immediately after injection. The volume of water represents the initial volume of distribution (V). It may be calculated easily:

image (1)

where C0 is the initial concentration. Therefore:

image (2)

A more accurate measurement of V is possible during constant rate infusion when the distribution of the drug in the tissues has time to equilibrate; this is termed volume of distribution at steady state (Vss).

Drugs which remain in the plasma and do not pass easily to other tissues have a small V and therefore a large C0. Relatively ionized drugs (e.g. muscle relaxants) or drugs highly bound to plasma proteins (e.g. NSAIDs) often have a small V. Drugs with a large V are often lipid-soluble and therefore penetrate and accumulate in tissues outside the plasma (e.g. intravenous induction agents). Some drugs accumulate outside the plasma, making values for V greater than total body volume (a reminder of the abstract nature of pharmacokinetics). Large V values are often observed for drugs highly bound to proteins outside plasma (e.g. local anaesthetics, digoxin).

Several factors may affect V and therefore C0 on bolus injection of a drug. Patients who are dehydrated, or have lost blood, have a significantly greater plasma C0 after a normal dose of intravenous induction agent, increasing the likelihood of severe side-effects, especially hypotension. Neonates have a proportionally greater volume of extracellular fluid compared with adults, and water-soluble drugs (e.g. muscle relaxants) tend to have a proportionally greater V. Factors affecting plasma protein binding (see above) may also affect V.

Finally, V can give some indication as to the half-life. A large V is often associated with a relatively slow decline in plasma concentration; this relationship is expressed below in a useful pharmacokinetic equation (eqn 4).

Clearance

Clearance is defined as the volume of blood or plasma from which the drug is removed completely in unit time. Drugs may be eliminated from the blood by the liver, kidney or occasionally other routes (see above). The relative proportion of hepatic and renal clearance of a drug is important. Most drugs used in anaesthetic practice are cleared predominantly by the liver, but some rely on renal or non-organ-dependent clearance. Excessive accumulation of a drug occurs in patients in renal failure if its renal clearance is significant. For example, morphine is metabolized primarily in the liver and this is not affected significantly in renal impairment. However, the active metabolite morphine-6-glucuronide is excreted predominantly by the kidney. This accumulates in renal insufficiency and is responsible for increased morphine sensitivity in these patients.

As with volume of distribution, clearance may suggest likely properties of a drug. For example, if clearance is greater than hepatic blood flow, factors other than hepatic metabolism must account for its total clearance. Values greater than cardiac output may indicate metabolism in the plasma (e.g. succinylcholine) or other tissues (e.g. remifentanil). Clearance is an important (but not the only) factor affecting t1/2 and steady-state plasma concentrations achieved during constant rate infusions (see below).

Elimination Half-Life

Methods of administration of a drug are influenced considerably by its plasma t1/2, as this often reflects duration of action. It is important to remember that t1/2 is influenced not only by clearance (Cl) but also by V:

image (3)

or

image

The constant in this equation (elimination rate constant) is the natural logarithm of 2 (ln 2) i.e. 0.693. Therefore:

image (4)

Half-life often reflects duration of action but not if the drug acts irreversibly (e.g. some NSAIDs, omeprazole, phenoxybenzamine) or if active metabolites are formed (Table 1.1).

So far, we have considered metabolic or elimination t1/2 only. The initial decrease in plasma concentrations after administration of many drugs, especially if given intravenously, occurs primarily because of redistribution into tissues. Therefore, the simple relationship between elimination t1/2 and duration of action does not apply in many situations (see below, ‘Two-Compartment Models’).

Calculating t1/2, V and Clearance

It is a simple exercise to calculate these values for a drug after intravenous bolus administration. A known dose is given and regular blood samples are taken for plasma concentration measurements. In this example, we assume that the drug remains in the plasma and is removed only by metabolism; this is a called a one-compartment model. After achieving C0, plasma concentration (CP) declines in a simple exponential manner as shown in Figure 1.5A. If the natural logs of the concentrations are plotted against time (semilog plot), a straight line is produced (Figure 1.5B). The gradient of this line is the elimination rate constant k, which is related to t1/2 in the following equation:

image (5)

We may calculate V using equation (2) and then clearance from equation (4). CP may be predicted at any time from the following equation:

image (6)

where t is the time after administration.

Clearance may be derived also by calculation of the area under the concentration-time curve extrapolated to infinity (AUC) and substitution in the following equation:

image (7)

Two-Compartment Models

The body is not, of course, a single homogeneous compartment; drug plasma concentrations are the result of elimination by metabolism and redistribution to and from tissues such as brain, heart, liver, muscles and fat. The mathematics describing this real situation are extremely complex. However, plasma concentrations of many drugs behave approximately as if they were distributed in two or three compartments. Applying these mathematical models is a reasonable compromise.

Let us consider a two-compartment model; one compartment may be thought of as representing the plasma and the other, the remainder of the body. When an intravenous bolus is injected into this system, CP decreases because of an exponential decay resulting from elimination and another exponential decay resulting from redistribution into the tissues. Therefore, when CP is plotted against time, the curve may be described by a biexponential equation. If plotted on a semilogarithmic plot (Fig. 1.6), two straight lines can be identified and derived. Their gradients are the elimination rate constants dependent on elimination (β) and redistribution (α).

Redistribution kinetics are not only of theoretical interest, because it is often the decline in CP resulting from redistribution which is responsible for the cessation of an observed effect of a drug; intravenous induction agents and initial doses of intravenous fentanyl are good examples of this. Patients wake up after a bolus administration of propofol because of redistribution, not metabolism.

Calculating the separate pharmacokinetic values is easy; one curve is simply subtracted from the other. Consider Figure 1.6 in which natural log concentration is plotted against time and two slopes are seen. The second and less steep slope represents decline in plasma concentration caused by elimination of the drug by metabolism. From this, the elimination half-life (t1/2β) may be calculated. In order to calculate the half-life of the redistribution phase (t1/2α), the elimination slope is extrapolated back to time 0. If data on this imaginary part of the elimination slope are subtracted from those on the real line above it, another imaginary line may be constructed which represents that part of the decline in plasma concentration which is the result of redistribution. From this line, the redistribution half-life (t1/2α) may be calculated.

The equation for CP at any time in a two-compartment model after bolus intravenous administration is therefore:

image (8)

where α and β are the redistribution and elimination rate constants, respectively, and A and B are values derived by back extrapolation of the redistribution and elimination slopes to the y-axis.

Some drugs, e.g. propofol, are best fitted to a triexponential, three-compartment model which reveals half-lives for two processes of redistribution (conventionally t1/2α and t1/2β) and one for elimination (t1/2γ). Equations developed from these basic concepts are contained in the software of target-controlled infusion pumps.

Context-Sensitive Half-Life

This concept refers to plasma half-life (time for plasma concentration to decline by 50%) after an intravenous drug infusion is stopped; ‘context’ refers to the duration of infusion. The amount of drug accumulating in body tissues increases with duration of infusion for most drugs. Consequently, on stopping the infusion, time for the plasma concentration to decline by 50% depends on duration of infusion. The longer the infusion, the more drug accumulates and the longer the plasma half-life becomes, because there is more drug to enter the plasma on stopping the infusion.

Figure 1.7 shows the effect of infusion duration on the half-lives of alfentanil, fentanyl and remifentanil. Alfentanil, and especially fentanyl, accumulate during infusion, causing an increase in context-sensitive half-life as the duration of infusion increases. In other words, time to recovery from alfentanil- or fentanyl-based anaesthesia depends on duration of infusion. Remifentanil is metabolized by tissue esterases and does not accumulate. Therefore, time for plasma concentration of remifentanil to decline by 50% is independent of duration of infusion, i.e. recovery times after remifentanil-based anaesthesia are short and predictable, no matter how long the infusion has run.

PHARMACOGENETICS

Pharmacogenetics refers to genetic differences in metabolic pathways which can affect individual responses to drugs, both in terms of therapeutic effect as well as adverse effects. Pharmacogenetics is a rising concern in clinical oncology, because the therapeutic window of most anticancer drugs is narrow and patients with impaired ability to detoxify drugs may undergo life-threatening toxicities.

The first observations of genetic variation in drug response date from the 1950s, involving the muscle relaxant (succinylcholine [suxamethonium]). Up to 4% of patients have a less efficient variant of the enzyme butyrylcholinesterase (plasma cholinesterase), which metabolizes succinylcholine chloride. As a consequence, the drug’s effect is prolonged to varying degrees (depending on the nature of the abnormal gene and whether the individual is homozygous or heterozygous), with slower recovery from paralysis (see Ch 6).

Variation in the N-acetyltransferase gene divides people into ‘slow acetylators’ and ‘fast acetylators’, with very different half-lives and blood concentrations of drugs such as isoniazid and procainamide.

As part of the inborn system for clearing the body of xenobiotics, the cytochrome P450 oxidases (CYPs) are heavily involved in drug metabolism, and genetic variations in CYPs affect large populations. One member of the CYP superfamily, CYP2D6, now has over 75 known allelic variations, some of which lead to no activity, and some to enhanced activity. An estimated 29% of people in some parts of the world may have multiple copies of the gene, and therefore may not be treated adequately with standard doses of drugs such as codeine (which is activated by the enzyme).

METHODS OF DRUG ADMINISTRATION

Oral

The oral route of drug administration is important in modern anaesthetic practice (e.g. premedication, postoperative analgesia). It is often necessary also to continue concurrent medication during the perioperative period (e.g. antihypertension therapy, anti-anginal medication). It is therefore important to appreciate the factors involved in the absorption of orally administered drugs.

The formulation of tablets or capsules is very precise, as their consistent dissolution is necessary before absorption can take place. The rate of absorption, and therefore effect of the drug, may be influenced significantly by this factor. Most preparations dissolve in the acidic gastric juices and the intact drug is absorbed in the upper intestine. However, some drugs are broken down by acids (e.g. omeprazole, benzylpenicillin) or are irritant to the stomach (e.g. aspirin, phenylbutazone) and may be given as enteric-coated preparations. Drugs given in solution are often absorbed more rapidly but this may induce nausea or vomiting immediately after anaesthesia. Some drugs used in anaesthetic practice are available in slow-release preparations (e.g. morphine, oxycontin, tramadol).

Intramuscular

Intramuscular administration is still used occasionally in the perioperative period. It may avoid the problems associated with large initial plasma concentrations after rapid intravenous administration, is devoid of first-pass effects and may be administered relatively easily. However, absorption may be unpredictable, some preparations are particularly painful and irritant (e.g. diclofenac) and complications include damage to nervous and vascular tissue and inadvertent intravenous injection. It is disliked intensely by most adults and nearly all children.

Variations in absorption may be clinically relevant. For example, peak plasma concentrations of morphine may occur at any time from 5 to 60 min after intramuscular administration, an important factor in the failure of this method to produce good reliable analgesia (see Ch 41).

Intravenous

Bolus

The majority of drugs used in anaesthetic practice are given intravenously as boluses and the pharmacokinetics are described in some detail above. The major disadvantage of this method is that dangerously high drug concentrations may occur readily, particularly with drugs of narrow therapeutic index and large interpatient pharmacodynamic and pharmacokinetic variations (i.e. most drugs used in anaesthetic practice). Therefore, it is an important general rule that all drugs administered intravenously should be given slowly. Manufacturers’ recommendations in this regard are often surprising; for example, a 10 mg dose of metoclopramide should be given over 1–2 min.

Only two factors have a major influence on the plasma concentrations achieved during a bolus intravenous injection: speed of injection and cardiac output. Therefore, an elderly, sick or hypovolaemic patient undergoing intravenous induction of anaesthesia is likely to suffer significant side-effects if the drug is given at the same rate as would be used in a normal, healthy young adult.

Infusion

Drugs may be given by constant-rate infusion, a method used frequently for propofol, neuromuscular blocking agents, opioids and many other drugs. Plasma concentrations achieved during infusions may be described by a simple wash-in exponential curve (Fig. 1.8). The only factor influencing time to reach steady-state concentration is t1/2. Maximum concentration is achieved after approximately 4–5 half-lives. Therefore, this method of administration is best suited to drugs with short half-lives, such as remifentanil, glyceryl trinitrate, Adrenaline and dopamine. However, in practice, it is often used for drugs such as morphine. Assuming a morphine t1/2 of 4 h, it will be about 20 h before steady-state concentration is reached (although an effective concentration can be achieved by initial bolus doses or a more rapid infusion). Therefore, vigilant observation is required with this method of delivery, especially if active metabolites are involved – in this example, morphine-6-glucuronide.

There is a simple equation describing the concentration achieved at steady state during a constant-rate infusion; this is based on the principle that, at steady state, the amount of drug cleared from the plasma is equal to that delivered:

image (9)

where Css is the concentration at steady state.

Many pathological conditions reduce drug clearance and may therefore result in unexpectedly large plasma concentrations during infusions. Half-life does not influence Css, only how quickly it is achieved.

Patient-Controlled Analgesia (PCA)

The use of PCA for the treatment of postoperative pain has become widespread and is described in detail in Chapter 41. The patient titrates opioid delivery to requirements by pressing a button on a PCA device which results in the delivery of a small bolus dose. A lockout time is set which does not allow another bolus to be delivered until the previous dose has had time to have an effect. There is an enormous interpatient variability in opioid requirement after surgery; effective and closely monitored PCA is able to cope with this.

Transdermal

Drugs with a high lipid solubility and potency may be given transdermally. The pharmacological properties of glyceryl trinitrate render it ideal for this technique (i.e. potent, highly lipid-soluble, short half-life). Transdermal hyoscine is used for travel and other causes of sickness. Fentanyl patches can be very effective, particularly in patients with cancer pain. Buprenorphine and lidocaine transdermal delivery systems are also available. The latter is used for post-herpetic neuralgia which has not responded to more conventional techniques.

It may take some time before a steady-state plasma concentration is achieved and many devices incorporate large amounts of drug in the adhesive layer in order to provide a loading dose which reduces this period. At steady state, transdermal delivery has several similarities to intravenous infusion. However, on removing the adhesive patch, plasma concentrations may decline relatively slowly because of a depot of drug in the surrounding skin; this occurs with transdermal fentanyl systems.

DRUG INTERACTIONS

There are three basic types of drug interaction; examples are listed in Table 1.2.

TABLE 1.2

Examples of Drug Interactions in Anaesthesia

Type Drugs Effect
Pharmaceutical Thiopental: succinylcholine Ampicillin: glucose, lactate
Blood: dextrans
Hydrolysis of succinylcholine Reduced potency

Rouleaux formation
Cross-matching difficulties

Plastic: glyceryl trinitrate Sevoflurane: soda lime Adsorption to plastic
Compound A
Pharmacokinetic Opioids: most drugs
Warfarin: NSAIDs
Barbiturates: warfarin
Neostigmine: succinylcholine
Delayed oral absorption
↑ Free warfarin
↑ Warfarin metabolism
↓ Succinylcholine metabolism
Pharmacodynamic Volatiles: opioids ↓ MAC
Volatiles: benzodiazepines ↓ MAC
Volatiles: N2O ↓ MAC
Volatiles: muscle relaxants ↑ Relaxation
Morphine: naloxone Reversal (receptor antagonism)
Muscle relaxants: neostigmine ↑ Relaxation

VOLATILE ANAESTHETIC AGENTS

Mechanism of Action

The exact mechanism of action of volatile anaesthetic agents is at present unknown. Potency is, in general, related to lipid solubility (Meyer-Overton relationship, Table 1.3) and this has given rise to the concept of volatile agents dissolving in the lipid cell membrane in a non-specific manner, disrupting membrane function and thereby influencing the function of proteins, e.g. ion channels. However, it is now appreciated that volatile agents affect neuronal function as a consequence of binding to specific protein sites (e.g. GABAA receptor).

Onset of Action

When considering onset of action of volatile agents, there is a fundamental difference compared with intravenous agents. Effects of non-volatile drugs are related to plasma or tissue concentrations; this is not so with volatile agents. Partial pressure of the volatile agent is important, not concentration. If a volatile agent is highly soluble in blood, partial pressure increases slowly because large amounts dissolve in the blood. Consequently, onset of anaesthesia is slow with agents soluble in blood and rapid with agents which are relatively insoluble. The same applies to recovery from anaesthesia. Table 1.4 lists the most commonly used inhaled agents (in order of speed of onset) and their relative blood/gas solubilities.

Alveolar partial pressure (PA) is assumed to be equivalent to cerebral artery partial pressure and therefore depth of anaesthesia. At a fixed inspired partial pressure (PI), the rate at which PA approaches PI is related to speed of onset of effect (Fig. 1.9). This is rapid with agents of low blood solubility (e.g. sevoflurane) and relatively slow with more soluble agents (e.g. halothane).

Clearly, solubility of the agent in blood is a major determinant of the speed of onset of anaesthesia, but other factors can have significant effects. The rate of delivery of the agent to the alveoli is important; therefore increasing PI by adjusting the vaporizer (a factor limited with some agents by irritant effects on the airway in spontaneously breathing patients), reducing apparatus dead space and increasing alveolar ventilation increase speed of induction of anaesthesia. If cardiac output is reduced, relatively less agent is removed from the alveolus and PA increases towards PI more rapidly. Consequently, induction of anaesthesia is more rapid in patients with reduced cardiac output. Both rate of delivery and cardiac output have particularly significant effects with agents that are relatively soluble in blood but less so with insoluble agents.

Ventilation/perfusion mismatch may reduce the speed of induction, an effect more significant in agents of low solubility. For example, if one lung is collapsed (i.e. perfused but not ventilated) increasing ventilation or inspired concentration of agents such as halothane helps to compensate. However, this is not the case for agents such as sevoflurane.