Drugs Acting on the Cardiovascular System

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 6095 times

Drugs Acting on the Cardiovascular System

Many drugs have either primary or secondary effects on the cardiovascular and autonomic nervous systems. Several of the drugs discussed in this chapter have more than one clinical indication and the drugs are considered according to their mechanism of action. An understanding of drugs acting on the cardiovascular and autonomic nervous systems requires an understanding of autonomic physiology and pharmacology.

THE AUTONOMIC NERVOUS SYSTEM

The term autonomic nervous system (ANS) refers to the nervous and humoral mechanisms which modify the function of the autonomous or automatic organs. These include heart rate and force of contraction, calibre of blood vessels, contraction and relaxation of smooth muscle in gut, bladder and bronchi, visual accommodation and pupillary size. Other functions include regulation of secretion from exocrine and other glands and aspects of metabolism (e.g. glycogenolysis and lipolysis) (Table 8.1). There is constant activity of both the sympathetic and parasympathetic nervous systems even at rest. This is termed sympathetic or parasympathetic tone and allows alterations in autonomic activity to produce rapid two-way regulation of physiological effect. The ANS is controlled by centres in the spinal cord, brainstem and hypothalamus, which are in turn influenced by higher centres in the cerebral and particularly the limbic cortex. The ANS is also influenced by visceral reflexes whereby afferent signals enter the autonomic ganglia, spinal cord, hypothalamus or brainstem and directly elicit appropriate reflex responses via the visceral organs. The efferent autonomic signals are transmitted through the body to two major subdivisions (separated by anatomical, physiological and pharmacological criteria), the sympathetic and the parasympathetic nervous systems.

The Sympathetic Nervous System

The sympathetic nervous system includes nerves which originate in the spinal cord between the first thoracic and second lumbar segments (T1 to L2). Fibres leave the spinal cord with the anterior nerve roots and then branch off as white rami communicantes to synapse in the bilateral paravertebral sympathetic ganglionic chains, although some preganglionic fibres synapse instead in the paravertebral ganglia (e.g. coeliac, mesenteric and hypogastric) in the abdomen before travelling to their effector organ with the relevant arteries. Postganglionic fibres travel from paravertebral ganglia in sympathetic nerves (to supply the internal viscera, including the heart) and spinal nerves (which innervate the peripheral vasculature and sweat glands). Sympathetic nerves throughout the circulation contain vasoconstrictor fibres, particularly in the kidneys, spleen, gut and skin; however, sympathetic vasodilator fibres predominate in skeletal muscle, and coronary and cerebral vessels. Sympathetic stimulation therefore causes predominantly vasoconstriction but also a redistribution of blood flow to skeletal muscle; constriction of venous capacitance vessels may decrease their volume and thereby increase venous return. The effects of sympathetic stimulation at different receptors and effector organs are summarized in Table 8.1. The distribution of sympathetic nerve fibres to an organ or region may differ from the sensory or motor supply, according to its embryonic origin. For example, sympathetic fibres to the heart arise from T1 to T5 (but predominantly from T1 to T4), the neck is supplied by fibres from T2, the chest by fibres from T3 to T6 and the abdomen by fibres from T7 to T11.

Sympathetic Neurotransmitters

The neurotransmitter present in preganglionic neurones is acetylcholine (ACh). These and other neurones containing ACh are termed cholinergic. However, the activity of preganglionic neurones is modulated by several other neuropeptides including enkephalin, neurotensin, substance P, somatostatin, nitric oxide, serotonin and catecholamines. ACh is the transmitter at all preganglionic synapses, acting via nicotinic receptors. Postganglionic sympathetic neurones secrete noradrenaline and are termed adrenergic (except for postganglionic sympathetic nerve fibres to sweat glands, pilo-erector muscles and some blood vessels, which are cholinergic).

Activation of preganglionic nicotinic fibres to the adrenal medulla causes the release of adrenaline (adrenaline), which is released primarily as a circulating hormone and is only found in insignificant amounts in the nerve endings. Endogenous catecholamines (adrenaline, noradrenaline (norepinephrine) and dopamine) are synthesized from the essential amino acid phenylalanine. Their structure is based on a catechol ring (i.e. a benzene ring with -OH groups in the 3 and 4 positions), and an ethylamine side chain (Figs 8.1 and 8.2); substitutions in the side chain produce the different compounds. Dopamine may act as a precursor for both adrenaline and noradrenaline when administered exogenously (see below).

The action of noradrenaline released from sympathetic nerve endings is terminated in one of three ways:

Most noradrenaline released from sympathetic nerves is taken back into the presynaptic nerve ending for storage and subsequent reuse. Re-uptake is by active transport back into the nerve terminal cytoplasm and then into cytoplasmic vesicles. This mechanism of presynaptic re-uptake, termed uptake1, is dependent on adenosine triphosphate (ATP) and Mg2 +, is enhanced by Li+ and may be blocked by cocaine and tricyclic antidepressants. Endogenous catecholamines entering the circulation by diffusion from sympathetic nerve endings or by release from the adrenal gland are metabolized rapidly by the enzymes monamine oxidase (MAO) and catechol O-methyltransferase (COMT) in the liver, kidneys, gut and many other tissues. The metabolites are conjugated before being excreted in urine as 3-methoxy-4-hydroxymandelic acid, metanephrine (from adrenaline) and normetanephrine (from noradrenaline) (Fig. 8.3). Noradrenaline taken up into the nerve terminal may also be deaminated by cytoplasmic MAO.

Another mechanism for the postsynaptic cellular re-uptake of catecholamines, termed uptake2, is present predominantly at the membrane of smooth muscle cells. It may be responsible for the termination of action of catecholamines released from the adrenal medulla.

Adrenergic Receptor Pharmacology

The actions of catecholamines are mediated by specific postsynaptic cell surface receptors. The original classification of these receptors into α- and β-adrenergic receptors was based upon the effects of adrenaline at peripheral sympathetic sites, α-receptors being responsible for vasoconstriction and β-receptors mediating effects on the heart, and bronchial and intestinal smooth muscle. However, several subtypes of α- and β-receptors exist in addition to receptors specific for dopamine (DA1 and DA2 subtypes). Two α- and β-receptor subtypes are well defined on functional, anatomical and pharmacological grounds (α1 and α2, β1 and β2). A third β-receptor subtype, β3, is found in adipocytes, skeletal and ventricular muscle, and the vasculature. At least three further subtypes of both α1– and α2-receptors and five subtypes of DA-receptor have also been identified, although their precise functions are unclear. Differentiation of receptor subtypes is now based more directly on the effects of various catecholamine agonist compounds (including endogenous catecholamines). Noradrenaline and adrenaline are agonists at both α1– and α2-receptors; noradrenaline is slightly more potent at α1-receptors, and more potent at α2-receptors. Adrenaline has a more potent action at β1-receptors, and also acts at β2-receptors, whereas noradrenaline has no β2-effects.

Until recently, it was thought that β1-receptors predominated in the heart, mediating increases in force and rate of contraction, and β2-receptors existed in bronchial, uterine and vascular smooth muscle, mediating relaxation. In fact, most organs and tissues contain both β1– and β2-receptors, which may even serve the same function. For example, up to 25% of cardiac β-receptors in the normal individual are of the β2 subtype, and this proportion may be increased in patients with heart failure. It is now apparent that β1-receptors in tissues are situated on the postsynaptic membrane of adrenergic neurones and respond to released noradrenaline. β2-Receptors are presynaptic and, when stimulated (principally by circulating catecholamines), they modulate autonomic activity by promoting neuronal noradrenaline release. β3-Receptors mediate thermogenesis and lipolysis in adipocytes but antagonize the effects of β1– and β2-receptors on the heart, and also mediate vasodilatation. Similarly, α1-receptors are present on the postsynaptic membrane, whereas α2-receptors are predominantly presynaptic, responding to circulating adrenaline but also mediating feedback inhibition of sympathetic nerve activity. Central α2 stimulation causes decreases in arterial pressure, peripheral resistance, venous return, myocardial contractility, cardiac output and heart rate by inhibition of sympathetic outflow. Postsynaptic α2-receptors present on platelets and in the CNS mediate platelet aggregation and membrane hyperpolarization, respectively.

Postsynaptic dopamine receptors (DA1) are present in vascular smooth muscle of the renal, splanchnic, coronary and cerebral circulations, where they mediate vasodilatation. They are also situated on renal tubules, where they inhibit sodium reabsorption, causing natriuresis and diuresis. Postsynaptic DA2-receptors are widespread in the CNS and occur on the presynaptic membrane of sympathetic nerves and in the adrenal gland. Stimulation of presynaptic DA2-receptors inhibits dopamine release by negative feedback.

Postganglionic sympathetic fibres supplying sweat glands and arterioles in some areas of skin and skeletal muscle are cholinergic. Vascular smooth muscle also contains non-innervated cholinergic receptors which mediate vasodilatation in response to circulating agonists. Cholinergic effects on vascular smooth muscle are usually minimal but may be involved in the mechanism of vasovagal attacks.

These subdivisions and the functions of the autonomic nervous system are summarized in Table 8.1.

Second and Third Messenger Systems

In addition to functional differences, α- and β-receptors differ in the intracellular mechanisms by which they act. Stimulation of β1– and β2-receptors activates Gs-proteins, which activate adenylate cyclase and cause the generation of intracellular cyclic adenosine monophosphate (cAMP). cAMP activates intracellular enzyme pathways (the third messengers) to produce the associated alteration in cell function (e.g. increased force of cardiac muscle contraction, liver glycogenolysis, bronchial smooth muscle relaxation). In cardiac myocytes, the intracellular pathway involves the activation of protein kinases to phosphorylate intracellular proteins and increase intracellular Ca2 + concentrations. Intracellular cAMP concentration is modulated by the enzyme phosphodiesterase, which breaks down cAMP to inactive 5′ AMP. This is the site of action of phosphodiesterase inhibitor drugs. The balance between production and degradation of cAMP is an important regulatory system for cell function. α2-Receptors interact with Gi-proteins to inhibit adenylate cyclase and Ca2 + channels, but activate K+ channels, phospholipase C and phospholipase A2. Cholinergic M2-receptors and somatostatin affect Gi-proteins in the same way.

In contrast, α1-receptor stimulation does not directly affect intracellular cAMP levels, but causes coupling with another G-protein, Gq, to activate membrane- bound phospholipase C. This in turn hydrolyses phosphatidylinositol biphosphate (PIP2) to inositol triphosphate (IP3), which produces changes in intracellular Ca2 + concentration and binding. These lead, for example, to smooth muscle contraction.

The Parasympathetic Nervous System

The parasympathetic nervous system controls vegetative functions, e.g. the digestion and absorption of nutrients, excretion of waste products and the conservation and restoration of energy. Parasympathetic neurones arise from cell bodies of the motor nuclei of cranial nerves III, VII, IX and X in the brainstem, and from the sacral segments of the spinal cord (‘the craniosacral outflow’). Preganglionic fibres run almost to the organ innervated and synapse in ganglia within the organ, giving rise to postganglionic fibres which then supply the relevant tissues. The ganglion cells may be well organized (e.g. the myenteric plexus of the intestine) or diffuse (e.g. in the bladder or vasculature). As the majority of all parasympathetic nerves are contained in branches of the vagus nerve, which innervates the viscera of the thorax and abdomen, increased parasympathetic activity is characterized by signs of vagal overactivity. Parasympathetic fibres also pass to the eye via the oculomotor (third cranial) nerve, and to the lacrimal, nasal and salivary glands via the facial (fifth) and glossopharyngeal (ninth) nerves. Fibres originating in the sacral portion of the spinal cord pass to the distal GI tract, bladder and reproductive organs. The effects of parasympathetic stimulation at different receptors and effector organs are summarized in Table 8.1

Parasympathetic Neurotransmitters

The chemical neurotransmitter at both pre- and postganglionic synapses is ACh, although transmission at postganglionic synapses may be modulated by other substances, including GABA, serotonin and opioid peptides. ACh is synthesized in the cytoplasm of cholinergic nerve terminals by the combination of choline and acetate (in the form of acetyl-CoA, which is synthesized in the mitochondria as a product of normal cellular metabolism). ACh is stored in specific agranular vesicles and released from the presynaptic terminal in response to neuronal depolarization to act at specific receptor sites on the postsynaptic membrane. It is rapidly metabolized by the enzyme acetylcholinesterase (AChE) to produce acetate and choline. Choline is then taken up into the presynaptic nerve ending for the regeneration of ACh. AChE is synthesized locally at cholinergic synapses, but is also present in erythrocytes and parts of the CNS. Butyryl cholinesterase (also termed plasma cholinesterase or pseudocholinesterase) is synthesized in the liver and is found in the plasma, skin, GI tract and parts of the CNS, but not at cholinergic synapses or the neuromuscular junction. It may metabolize ACh, in addition to some neuromuscular blockers (e.g. succinylcholine and mivacurium), but its physiological role probably involves the breakdown of other choline esters which may be present in the intestine.

Parasympathetic Receptor Pharmacology

Parasympathetic receptors have been classified according to the actions of the alkaloids muscarine and nicotine. The actions of ACh at the postganglionic membrane are mimicked by muscarine and are termed muscarinic, whereas preganglionic transmission is termed nicotinic. ACh is also the neurotransmitter at the neuromuscular junction, via nicotinic receptor sites. Five subtypes of muscarinic receptors (M1–M5) have been characterized; all five subtypes exist in the CNS, but there are differences in their peripheral distribution and function (Table 8.2). M1-receptors are found in the stomach, where they mediate acid secretion, and in inflammatory cells in the lung (including mast cells and eosinophils) where they may have a role in airway inflammation. M2-receptors predominate in the myocardium, where they modulate heart rate and impulse conduction. Prejunctional M2-receptors are also involved in the regulation of synaptic noradrenaline and postganglionic ACh release. M3-receptors are present in classic postsynaptic sites in glandular tissue (of the GI and respiratory tract) and bronchial smooth muscle, where they mediate most of the post-junctional effects of ACh. M4-receptors have been isolated in cardiac and lung tissue in animal models and may have inhibitory effects, but the distribution and functions of M5-receptors are not yet defined. In common with adrenergic receptors, muscarinic receptors are coupled to membrane-bound G-proteins although the subtypes differ in the second messenger system with which they interact. Currently available anticholinergics probably act at all muscarinic receptor subtypes but their clinical spectra differ, which suggests that they may have differential effects at different subtypes.

DRUGS ACTING ON THE SYMPATHETIC NERVOUS SYSTEM

Sympathomimetic Drugs

Sympathomimetic drugs partially or completely mimic the effects of sympathetic nerve stimulation or adrenal medullary discharge. They may act:

The drugs may be classified according to their structure (catecholamine/non-catecholamine), their origin (endogenous/synthetic) and their mechanism of action (via adrenergic receptors or via a non-adrenergic mechanism) (Table 8.3). Drugs which affect myocardial contractility are termed inotropes, although this term is usually applied to those drugs that increase cardiac contractility (strictly ‘positive inotropes’). Myocardial contractility may be increased by:

Inotropes may also be classified into positive inotropic drugs which also produce systemic vasoconstriction (‘inoconstrictors’) and those which also produce systemic vasodilatation (‘inodilators’). Inoconstrictors include noradrenaline, adrenaline and ephedrine. Inodilators are dobutamine, dopexamine, isoprenaline and phosphodiesterase inhibitors. Dopamine is an inodilator at low dose, and an inoconstrictor at higher doses.

Catecholamines

Catecholamine drugs may be endogenous (adrenaline, noradrenaline and dopamine) or synthetic (dobutamine, dopexamine and isoprenaline). Several other drugs with a non-catecholamine structure produce sympathomimetic effects via adrenergic receptors, e.g. ephedrine and phenylephrine. All catecholamine drugs are inactivated in the gut by MAO and are usually only administered parenterally. They all have very short half-lives in vivo, and so when given by intravenous infusion, their effects may be controlled by altering the infusion rate. The comparative effects of different inotropes and vasopressors are outlined below.

Endogenous Catecholamines

Adrenaline: Adrenaline comprises 80–90% of adrenal medullary catecholamine content and is also an important CNS neurotransmitter. It is a powerful agonist at both α- and β-adrenergic receptors, being slightly less potent than noradrenaline at α1-receptors but more potent at β-receptors. It is the treatment of choice in acute allergic (anaphylactic) reactions and is used in the management of cardiac arrest and shock, and occasionally as a bronchodilator. Except in emergency situations, i.v. injection is avoided because of the risk of inducing cardiac arrhythmias. Subcutaneous administration produces local vasoconstriction and so smoothes out its own effect by slowing absorption.

The effects of adrenaline on arterial pressure and cardiac output are dose-dependent. Although both α- and β-receptors are stimulated, β2-vasodilator effects are most sensitive. β1-Mediated effects cause marked increases in heart rate and contractility, cardiac output and systolic pressure. In low dosage, vasodilatation in skeletal muscle and splanchnic arterioles (β2) may predominate over α-mediated vasoconstriction in skin and renal vasculature; systemic vascular resistance and diastolic pressure may decrease, pulse pressure widens, but mean arterial pressure remains stable. At higher doses, α-mediated vasoconstriction becomes more prominent in venous capacitance vessels (increasing venous return) and the precapillary resistance vessels of skin, mucosa and kidney (increasing peripheral resistance). Systolic pressure increases further, but cardiac output may decrease. Adrenaline causes marked decreases in renal blood flow, but coronary blood flow is increased. In contrast to other sympathomimetics, adrenaline has significant metabolic effects. Hepatic glycogenolysis and lipolysis in adipose tissue increase (β1 and β3 effects), and insulin secretion is inhibited (α1 effect) so that hyperglycaemia occurs.

Adrenaline 0.5–1 mg i.m. (0.5–1.0 mL of 1:1000 solution) or 100 μg increments i.v. to a dose of 1 mg (1–10 mL of a 1:10 000 solution) is used to treat acute anaphylactic reactions. Adrenaline is important in the management of cardiac arrest (in doses of 1 mg i.v., repeated every 3–5 min), mostly because of its α effects; widespread systemic vasoconstriction occurs, increasing aortic diastolic pressure, and coronary and cerebral perfusion. Pure α-agonists are less effective than adrenaline in the management of cardiac arrest, and the β2 effects of adrenaline may contribute to improved cerebral perfusion. In emergency situations, it may also be administered via the tracheal route, in doses of 2–3 mg diluted to a volume of 10 mL. It is effective by aerosol inhalation in bronchial asthma but has been superseded by selective β2-agonists (see below). Unlike indirect-acting sympathomimetics which cause release of noradrenaline, tachyphylaxis should not occur with adrenaline. Adrenaline is also used as a topical vasoconstrictor to aid haemostasis and is incorporated into local anaesthetic solutions to decrease systemic absorption and prolong the duration of local anaesthesia.

Noradrenaline: Noradrenaline acts as a potent arteriolar and venous vasoconstrictor, acting predominantly at α-receptors, with a slightly greater potency there than adrenaline. It is also an agonist at β-receptors, but β2 effects are not apparent in clinical use. Infusions of noradrenaline increase venous return, systolic and diastolic systemic and pulmonary arterial pressures, and central venous pressure. Cardiac output increases but heart rate decreases because of baroreflex activity. At higher doses, the α-mediated effects of widespread intense vasoconstriction overcome β1 effects on cardiac contractility, leading to a decrease in cardiac output at the cost of increased myocardial oxygen demand in conjunction with reductions in renal blood flow and glomerular filtration rate. Its principal use is in the management of septic shock when systemic vascular resistance is low.

Dopamine: Dopamine is the natural precursor of adrenaline and noradrenaline. It stimulates both α- and β-adrenergic receptors in addition to specific dopamine DA1-receptors in renal and mesenteric arteries. Dopamine has a direct positive inotropic action on the myocardium via β-receptors and also by release of noradrenaline from adrenergic nerve terminals. The overall effects of dopamine are highly dose-dependent. In low dosage (< 3 μg kg–1 min–1), renal and mesenteric vascular resistances are reduced by an action on DA1-receptors, resulting in increased splanchnic and renal blood flows, glomerular filtration rate and sodium excretion. At doses of 5–10 μg kg–1 min–1, the increasing direct β-mediated inotropic action predominates, increasing cardiac output and systolic pressure with little effect on diastolic pressure; peripheral resistance is usually unchanged. At doses > 15 μg kg–1 min–1, α-receptor activity predominates, with direct vasoconstriction and increased cardiac stimulation (similar to noradrenaline). Renal and splanchnic blood flows decrease, and arrhythmias may occur. Dopamine receptors are widely present in the CNS, particularly in the basal ganglia, pituitary (where they mediate prolactin secretion) and the chemoreceptor trigger zone on the floor of the fourth ventricle (where they mediate nausea and vomiting). Recently, dopamine infusions have been associated with decreased prolactin secretion, and the use of ‘prophylactic’ dopamine infusions in an attempt to preserve renal function in perioperative or critically ill patients has declined.

Synthetic Catecholamines

Isoprenaline: Isoprenaline is a potent β1– and β2-agonist, with virtually no activity at α-receptors. It acts via cardiac β1-receptors, and at β2-receptors in the smooth muscle of bronchi, the vasculature of skeletal muscle and the gut. After intravenous infusion, heart rate increases and peripheral resistance is reduced. Cardiac output may increase because of increased heart rate and contractility but effects on arterial pressure are variable. Isoprenaline also reduces coronary perfusion pressure, increases myocardial oxygen consumption and causes arrhythmias. Other β2-mediated effects include relaxation of bronchial smooth muscle and stabilization of mast cells. It has been superseded for use in the treatment of severe asthma by newer specific β2-agonists with fewer cardiac effects. Its current indication is as an infusion in the treatment of bradyarrhythmias or atrioventricular heart block associated with low cardiac output (e.g. following acute myocardial infarction) because it increases heart rate and conduction by a direct action on the subsidiary pacemaker. This indication is usually an interim measure before insertion of a temporary pacing wire.

Dobutamine: Dobutamine is primarily a β1-agonist, with moderate β2– and mild α1-agonist activity, and no action at DA-receptors. Its primary effect is an increase in cardiac output via increased contractility (β1 effect) augmented by a reduction in afterload. Heart rate also increases (β2 effect). Systolic arterial pressure may increase but peripheral resistance is reduced or unchanged. There is no direct effect on venous tone or renal blood flow but preload may decrease and urine output and sodium excretion increase as a consequence of the increased cardiac output. Dobutamine increases SA node automaticity and conduction velocity in the atria, ventricles and AV node, but to a lesser extent than isoprenaline. Dobutamine infusion produces a progressive increase in cardiac output which is greater than with comparable doses of dopamine, although arterial pressure may remain unchanged. At higher doses, tachycardia and arrhythmias may occur, but dobutamine has less effect on myocardial oxygen consumption compared with other catecholamines. Dobutamine is widely used to optimize cardiac output in septic shock, often in combination with noradrenaline. It is also used alone or in combination with vasodilator drugs in heart failure when peripheral resistance is high, and to increase heart rate and cardiac output in myocardial stress testing.

Dopexamine: Dopexamine is a synthetic dopamine analogue which is an agonist at β2– and DA1-receptors. It is also a weak DA2-agonist and it inhibits the neuronal re-uptake of noradrenaline (uptake1), but has no direct effects at β1– or α-receptors. Its principal effect is β2-agonism, producing vasodilatation in skeletal muscle; it is less potent at DA1-receptors, but a more potent β2-agonist than dopamine. It produces mild increases in heart rate, contractility and cardiac output (effects on β2-receptors and noradrenaline uptake), renal and mesenteric vasodilatation (β2 and DA1 effects), and natriuresis (DA1 effect). Coronary and cerebral blood flows are also increased. Systemic vascular resistance decreases and arterial pressure may decrease if intravascular volume is not maintained. Dopexamine has theoretical advantages in maintaining cardiac output and splanchnic blood flow in patients with systemic sepsis or heart failure. It is also used for this purpose in patients undergoing major abdominal surgery. Dopexamine also has anti-inflammatory effects (in common with other β-agonists) which are independent of its effects on gut mucosal perfusion. It is metabolized by hepatic methylation and conjugation and is eliminated mostly via the kidneys.

Fenoldopam is a dopamine (DA1) agonist available in the USA which causes peripheral vasodilatation and increases renal blood flow and sodium and water excretion. It has been used in the treatment of hypertensive emergencies. Unlike some other vasodilators (e.g. sodium nitroprusside) it does not cause rebound hypertension after stopping the infusion.

Non-Catecholamine Sympathomimetics

Synthetic sympathomimetic drugs may mimic the effect of adrenaline at adrenergic receptors (direct-acting) or may produce effects by causing release of endogenous noradrenaline from postganglionic sympathetic nerve terminals (indirect-acting). Some drugs have direct and indirect sympathomimetic effects (e.g. ephedrine, metaraminol). Direct-acting compounds may affect α- or β-receptors selectively, whereas indirect-acting compounds have predominantly α- and β1-agonist effects (as noradrenaline is only a weak β2-agonist). Indirect-acting compounds are taken up into the nerve terminal via the noradrenaline re-uptake pathway, and so their effect is reduced by drugs which block noradrenaline re-uptake (e.g. tricyclic antidepressants). Conversely, the effect of direct-acting drugs is enhanced. In patients treated with drugs which decrease sympathetic nervous system activity (e.g. clonidine, reserpine), the cardiovascular response to indirect-acting drugs is diminished; however, upregulation of adrenergic receptors occurs and an increased response to direct-acting sympathomimetics is seen. Drugs with selective α-adrenergic receptor effects (e.g. phenylephrine, methoxamine) are potent vasoconstrictors.

Ephedrine: Ephedrine is a naturally occurring sympathomimetic amine which is now produced synthetically. It acts directly and indirectly as an agonist at α-, β1– and β2-receptors. The indirect actions are increased endogenous noradrenaline release and inhibition of MAO. Its cardiovascular effects are similar to those of adrenaline, but the duration of action is up to 10 times longer. It causes increases in heart rate, contractility, cardiac output and arterial pressure (systolic >  diastolic). It may predispose to arrhythmias. Systemic vascular resistance is usually unchanged because α-mediated vasoconstriction in some vascular beds is balanced by β-mediated vasodilatation in others, but renal and splanchnic blood flows decrease. It relaxes bronchial and other smooth muscle, and is occasionally used as a bronchodilator. It is active orally because it is not metabolized by MAO in the gut, and is useful by intramuscular injection because muscle blood flow is preserved. Ephedrine undergoes hepatic deamination and conjugation but significant amounts are excreted unchanged in urine. This accounts for its long duration of action and elimination half-life (3–6 h). Tachyphylaxis (a decreased response to repeated doses of the drug) occurs because of persistent occupation of adrenergic receptors and depletion of noradrenaline stores.

Ephedrine is often used to prevent or treat hypotension resulting from sympathetic blockade during regional anaesthesia or from the effects of general anaesthesia. Although widely used to prevent hypotension during regional anaesthesia in obstetric patients, it has largely been superseded for this indication by an infusion of phenylephrine, which has better effects on maternal and fetal haemodynamics. Oral or topical ephedrine is also useful as a nasal decongestant.

Phenylephrine: Phenylephrine is a potent synthetic direct-acting α1-agonist, which has minimal agonist effects at α2-and β-receptors. It has effects similar to those of noradrenaline, causing widespread vasoconstriction, increased arterial pressure, bradycardia (as a result of baroreflex activation) and a decrease in cardiac output. Venoconstriction predominates and diastolic pressure increases more than systolic pressure, so that coronary blood flow may increase. It is used as intermittent boluses (50-100 μg by slow injection) or an infusion (50–150 μg min− 1) to maintain arterial pressure during general or regional anaesthesia, and also topically as a nasal decongestant and mydriatic. Absorption of phenylephrine from mucous membranes may occasionally produce systemic side-effects.

Vasopressin: Arginine vasopressin (AVP) (formerly termed antidiuretic hormone) is a peptide hormone secreted by the hypothalamus. Its primary role is the regulation of body fluid balance. It is secreted in response to hypotension and promotes retention of water by action on specific cAMP-coupled V2-receptors. It causes vasoconstriction by stimulating V1-receptors in vascular smooth muscle and is particularly potent in hypotensive patients. It is increasingly used in the treatment of refractory vasodilatory shock which is resistant to catecholamines, although it can cause peripheral or splanchnic ischaemia. The vasopressin analogue desmopressin is used to treat diabetes insipidus. Another analogue, terlipressin, is used to limit bleeding from oesophageal varices in patients with portal hypertension as an adjunct to definitive treatment.

Phosphodiesterase Inhibitors: Phosphodiesterase inhibitors increase intracellular cAMP concentrations by inhibition of the enzyme responsible for cAMP breakdown (Fig. 8.4). Increased intracellular cAMP concentrations promote the activation of protein kinases, which lead to an increase in intracellular Ca2 +. In cardiac muscle cells, this causes a positive inotropic effect and also facilitates diastolic relaxation and cardiac filling (termed ‘positive lusitropy’). In vascular smooth muscle, increased cAMP decreases intracellular Ca2 + and causes marked vasodilatation. Several subtypes of phosphodiesterase (PDE) isoenzyme exist in different tissues. Theophylline is a non-specific PDE inhibitor, but the newer drugs (e.g. enoximone and milrinone) are selective for the PDE type III isoenzyme present in the myocardium, vascular smooth muscle and platelets. PDE III inhibitors are positive inotropes and potent arterial, coronary and venodilators. They decrease preload, afterload, pulmonary vascular resistance and pulmonary capillary wedge pressure (PCWP), and increase cardiac index. Heart rate may increase or remain unchanged. In contrast to sympathomimetics, they improve myocardial function without increasing oxygen demand or causing tachyphylaxis. Their effects are augmented by the co-administration of β1-agonists (i.e. increases in cAMP production are synergistic with decreased cAMP breakdown). They have particular advantages in patients with chronic heart failure, in whom downregulation of myocardial β-adrenergic receptors occurs, so that there is a decreased inotropic response to β-sympathomimetic drugs. A similar phenomenon occurs with advanced age, prolonged (> 72 h) catecholamine therapy and possibly with surgical stress.

PDE III inhibitors are indicated for acute refractory heart failure, e.g. cardiogenic shock, or pre- or postcardiac surgery. However, long-term oral treatment is associated with increased mortality in patients with congestive heart failure. All may cause hypotension, and tachyarrhythmias may occur. Other adverse effects include nausea, vomiting and fever. Their half-life is prolonged markedly in patients with heart or renal failure and they are commonly administered as an i.v. loading dose over 5 min with or without a subsequent i.v. infusion. Milrinone is a bipyridine derivative whereas enoximone is an imidazole derivative. Enoximone undergoes substantial first-pass metabolism, and is rapidly metabolized to an active sulphoxide metabolite which is excreted via the kidneys and which may accumulate in renal failure. The elimination t½ of enoximone is 1–2 h in healthy individuals but up to 20 h in patients with heart failure.

Calcium: Calcium ions are involved in cellular excitation, excitation-contraction coupling and muscle contraction in cardiac, skeletal and smooth muscle cells. Increased extracellular Ca2 + increases intracellular Ca2 + concentrations and consequently the force of contraction of cardiac myocytes and vascular smooth muscle cells. Massive blood loss and replacement with large volumes of calcium-free fluids or citrated blood (which chelates Ca2 +) may cause a decrease in serum Ca2 + concentration, especially in the critically ill. Therefore, Ca2 + salts (e.g. calcium chloride or gluconate) may be administered, particularly during and after cardiopulmonary bypass. Intravenous calcium 5 mg kg–1 may increase mean arterial pressure, but the effects on cardiac output and systemic vascular resistance are variable and there is little good evidence for the efficacy of Ca2 + salts. Moreover, high Ca2 + concentrations may cause cardiac arrhythmias and vasoconstriction, may be cytotoxic and may worsen the cellular effects of ischaemia. Calcium salts may be indicated for the treatment of hypocalcaemia (ionized Ca2 + < 0.8 mmol L–1), hyperkalaemia and calcium channel blocker toxicity.

Selective β2-Agonists

Selective β2-receptor agonists (e.g. salbutamol, terbutaline, formoterol and salmeterol) relax bronchial, uterine and vascular smooth muscle while having much less effect on the heart than isoprenaline. These drugs are partial agonists (their maximal effect at β2-receptors is less than that of isoprenaline) and are only partially selective for β2-receptors. They are used widely in the treatment of bronchospasm (see Ch 9). Although less cardiotoxic than isoprenaline, dose-related tremor, tachyarrhythmias, hyperglycaemia, hypokalaemia and hypomagnesaemia may occur. β2-Agonists are resistant to metabolism by COMT and therefore have a prolonged duration of action (mostly 3–5 h). Salmeterol is highly lipophilic, has a strong affinity for the β2-adrenergic receptor, is longer acting than the other β2-agonists and so is used for maintenance therapy in chronic asthma in combination with inhaled steroids. β2-Agonists are usually administered by the inhaled (metered dose inhaler or nebulizer) or intravenous routes because of unpredictable oral absorption and a high hepatic extraction ratio. When inhaled, only 10–20% of the administered dose reaches the lower airways; this proportion is reduced further when administered via a tracheal tube. Nevertheless, systemic absorption does occur, although adverse effects are less common during long-term therapy.

Sympatholytic Drugs

Sympatholytic drugs antagonize the effects of the sympathetic nervous system either at central adrenergic neurones, peripheral autonomic ganglia or neurones, or at postsynaptic α- or β-receptors. Most are hypotensive drugs, although they have other effects and indications.

Centrally Acting Sympatholytic Drugs

Centrally acting drugs act by stimulation of central α2-receptors to decrease sympathetic tone. They were used as antihypertensive drugs, but have been superseded for this purpose by newer drugs with fewer adverse effects. They are also agonists at central imidazoline (I1) receptors, which contributes to their hypotensive action. I1 receptors are present in several peripheral tissues, including the kidney. Central α2-stimulation causes decreases in arterial pressure, peripheral resistance, venous return, myocardial contractility, cardiac output and heart rate, but baroreceptor reflexes are preserved and the pressor response to ephedrine or phenylephrine may be exaggerated. Stimulation of peripheral α2-receptors on vascular smooth muscle causes direct arteriolar vasoconstriction, although the central effects of these drugs predominate overall. However, severe rebound hypertension may occur on stopping chronic oral therapy. α2-Receptors in the dorsal horn of the spinal cord modulate upward transmission of nociceptive signals by modifying local release of substance P and CGRP. Centrally acting α2-agonists produce analgesia by activation of descending spinal and supraspinal inhibitory pathways, and clonidine is now used mostly for its analgesic effects. These are greatest when administered by the epidural or spinal route. Other effects include dry mouth, sedation and anxiolysis.

Clonidine is a partial agonist at central and peripheral α2-receptors, and a central imidazoline (I1) receptor agonist. Clonidine has some effects at α1-receptors (α21 > 200:1); dexmedetomidine and azepexole are more α2-selective alternatives. Transient hypertension and bradycardia may occur after i.v. injection, caused by direct stimulation of peripheral vascular α2-receptors though an α1-agonist effect may also contribute. Clonidine potentiates the MAC of inhalational anaesthetic agents by up to 50%. It has a synergistic analgesic effect with opioids which may be partly pharmacokinetic because the elimination half-life of opioids is also increased. Clonidine is well absorbed orally, with peak plasma concentrations after 60–90 min. It is highly lipid-soluble and approximately 50% is metabolized in the liver to inactive metabolites; the rest is excreted unchanged via the kidneys, with an elimination half-life of 9–12 h. Clonidine 5 μg kg–1 as premedication attenuates reflex sympathetic responses and may reduce cardiac complications after non-cardiac surgery in patients at high risk of cardiovascular events. It is also used in the treatment of opioid withdrawal and postoperative shivering. Epidural clonidine 1–2 μg kg–1 increases the duration and potency of analgesia provided by epidural opioid or local anaesthetic drugs. α2-Agonists also have some antiarrhythmic effects, decreasing both the incidence of catecholamine-related arrhythmias and the toxicity of bupivacaine and cocaine.

Methyldopa crosses the blood-brain barrier easily and is converted to α-methyl noradrenaline, the active molecule, which is a full agonist at α2-receptors (α21 selectivity = 10:1). Adverse effects include peripheral oedema, hepatotoxicity, depression and a positive direct Coombs’ test; some patients develop haemolytic anaemia. Its use is largely restricted to the management of pregnancy-associated hypertension.

Moxonidine is a moderately selective imidazoline I1-receptor agonist (I1 > α2) which reduces central sympathetic activity by stimulation of medullary I1-receptors. It is used in the treatment of hypertension. Systemic vascular resistance is reduced but heart rate and stroke volume are unchanged. Moxonidine has few α2-related adverse effects but it may potentiate bradycardia and is contraindicated in sinoatrial block, and second- or third-degree AV block.

Peripherally Acting Sympatholytic Drugs

Adrenergic Neurone Blocking Drugs: Guanethidine decreases peripheral sympathetic nervous system activity by competitively binding to noradrenaline binding sites in storage vesicles in the cytoplasm of postganglionic sympathetic nerve terminals. Further uptake of noradrenaline into the vesicles is inhibited and it is metabolized by cytoplasmic MAO, so the nerve terminals become depleted of noradrenaline. Guanethidine has local anaesthetic properties and does not cross the blood–brain barrier. It is sometimes used to produce intravenous regional sympathetic blockade in the treatment of chronic limb pain associated with excessive autonomic activity (reflex sympathetic dystrophy or complex regional pain syndromes). Bretylium has a similar mode of action; it is used in the treatment of resistant ventricular arrhythmias (see below).

α-Adrenergic Receptor Antagonists: α-Adrenergic antagonists (α-blockers) selectively inhibit the action of catecholamines at α-adrenergic receptors. They are used mainly as vasodilators for the second-line treatment of hypertension or as urinary tract smooth muscle relaxants in patients with benign prostatic hyperplasia. They also have an important role in the preoperative management of phaeochromocytoma (see Ch 37).

α-Blockers diminish vasoconstrictor tone, causing venous pooling and a decrease in peripheral vascular resistance. In common with other vasodilators, they may have indirect positive inotropic actions as a result of reductions in afterload and preload, so cardiac output may increase. They may be classified according to their relative selectivity for α1– and α2-receptors. Non-selective α-blockers commonly induce postural hypotension and reflex tachycardia, partly because α2-blockade prevents the feedback inhibition of noradrenaline on its own release at presynaptic α2-receptors, and neuronal noradrenaline concentrations increase. The action of noradrenaline at cardiac β-receptors then limits the hypotensive effects of non-selective α-blockers. In addition, the proportions of pre- and postsynaptic α2-receptors in the arterial and venous smooth muscle may differ, so that α1-selective drugs have a more balanced effect on venous and arterial circulations. The co-administration of a β-blocker may attenuate reflex tachycardia and produces a synergistic effect on arterial pressure.

α1-Selective Antagonists: Selective α1-blockers include prazosin, doxazosin, indoramin, and urapidil. Doxazosin has largely succeeded prazosin as it has a more prolonged duration of action. Reflex tachycardia and postural hypotension are less common than with direct-acting vasodilators (e.g. hydralazine) and the non-selective α-blockers, but may still occur on initiating therapy. Nasal congestion, sedation and inhibition of ejaculation may occur.

Labetalol (see below) is a competitive α1-, β1– and β2-antagonist, which is more active at β- than at α-receptors. At low doses (5–10 mg i.v.), it decreases arterial pressure without producing a tachycardia. At higher doses, the β effect becomes more prominent, with negative inotropic and chronotropic effects. Carvedilol is an α1– and β-receptor antagonist which also has direct vasodilator effects (see below).

β-Adrenergic Receptor Antagonists

β-adrenergic receptor antagonists (β-blockers) are structurally similar to the β-agonists, e.g. isoprenaline. Variations in the molecular structure (primarily of the catechol ring) have produced compounds which do not activate adenylate cyclase and the second messenger system despite binding avidly to the β-adrenergic receptor. Most are stereoisomers and the L-form is generally more potent (as an agonist or antagonist) than the D-form. β-Blockers are competitive antagonists with high receptor affinity, although their effects are attenuated by high concentrations of endogenous or exogenous agonists. They may be classified according to:

β1– or β2-Adrenergic Receptor Affinity: The relative potency of β-blockers is less important than their relative effects on the different β-receptor subtypes. Compounds are available which block preferentially either β1– or β2-receptors, although in clinical practice the β1-selective drugs are more important. The first generation of β-blockers (e.g. propranolol, timolol) were non-selective; second-generation drugs (e.g. atenolol, metoprolol, bisoprolol) are selective for β1-receptors but have no ancillary effects. The third generation of β-blockers are β1-selective, but also have effects on other receptors (e.g. labetalol and carvedilol are antagonists at α1-adrenergic receptors, and celiprolol produces vasodilatation by a mechanism involving endothelial nitric oxide). β1-Selective (or ‘cardioselective’) drugs have theoretical advantages because some of the adverse effects of β-blockers are related to β2-antagonism, but the selectivity of both drugs and tissues is only relative: all β1-selective drugs antagonize β2-receptors at higher doses, and 25% of cardiac β-receptors are of the β2 subtype. However β1-selective drugs appear to have fewer adverse effects on blood glucose control in diabetics, less effect on serum lipids and less effect on bronchial tone in patients with chronic obstructive pulmonary disease.

Pharmacological Properties of β-Blockers: The pharmacological properties of β-blockers are summarized in Table 8.4. All are weak bases and most are well absorbed to produce peak plasma concentrations 1–3 h after oral administration. The more lipid-soluble drugs are almost completely absorbed, but are metabolized to a greater extent and tend to have a marked first-pass effect through the liver. This reduces their bioavailability, but is offset by the fact that the 4-hydroxylated metabolites so formed are also active. These active metabolites are excreted via the kidneys and may accumulate in patients with renal failure. Propranolol decreases both the clearance of amide local anaesthetics (by decreasing hepatic blood flow and inhibiting metabolism) and the pulmonary first-pass uptake of fentanyl. The first-pass metabolic pathways may also become saturated, so that proportionately higher plasma concentrations of the parent drug occur at higher oral doses. The first-pass effect is also a source of wide interindividual variation in plasma concentrations achieved from the same dose of primarily metabolized drugs, although β-blockers have a flat dose-response curve and large changes in plasma concentration may give rise to only a small change in degree of β-blockade. However, differences in individual plasma concentration–response relationships may occur, possibly as a result of variations in sympathetic tone or the formation of active metabolites. All β-blockers are distributed widely throughout the body and significant concentrations occur in the CNS, particularly for the more lipid-soluble drugs (e.g. propranolol).

The less lipid-soluble drugs (e.g. atenolol) are less well absorbed, are metabolized to a lesser extent, are excreted via the kidneys and tend to have longer half-lives. Atenolol, nadolol and sotalol are excreted largely unchanged in urine and so are little affected by impairment of liver function.

Indications for β-Blockade: See Tables 8.5 and 8.6 for indications for β-blockade.

TABLE 8.5

Clinical Indications for β-Blockade

Hypertension

Ischaemic heart disease

Secondary prevention of myocardial infarction

Obstructive cardiomyopathy

Congestive heart failure

Arrhythmias

Miscellaneous

TABLE 8.6

Specific Perioperative Indications for β-Blockers

Prevention or treatment of intraoperative hypertension, tachycardia and supraventricular tachyarrhythmias associated with excessive sympathetic activity

Treatment of postoperative hypertension

Controlled hypotension

Prophylaxis or treatment of perioperative myocardial ischaemia

Pre- and perioperative management of phaeochromocytoma

Pre- and perioperative management of thyrotoxic patients

Hypertension. β-Blockers have been regarded as first-line therapy for the treatment of hypertension for many years, either alone or in combination with other drugs. They are of proven benefit in reducing the incidence of stroke and the morbidity and mortality from coronary artery disease in younger hypertensive patients, although the effectiveness of β-blockade in patients aged > 60 years has been questioned recently. The antihypertensive effect results from a combination of factors.

image Reductions in heart rate, cardiac output and myocardial contractility.

image A reduction in central sympathetic nervous activity. The significance of this is uncertain because different drugs vary widely in their CNS penetration, but have similar effects on arterial pressure.

image Decreased plasma renin concentration. β-Blockers decrease resting and orthostatic release of renin to a variable extent. The non-selective drugs propranolol and timolol cause the greatest reduction, while partial agonists (oxprenolol, pindolol) or β1-selective drugs are less effective. However, no correlation has been found between renin-lowering effect and antihypertensive activity or dosage of β-blocker used.

image Effects on peripheral resistance. β-Blockade does not reduce peripheral resistance directly and may even cause an increase by allowing unopposed α-stimulation. As the vasodilating effect of catecholamines on skeletal muscle is β2-mediated, unopposed α stimulation would be expected to be lower with cardioselective drugs or partial agonists. However, cardioselectivity decreases with dosage and, because hypertensive patients often require a large dose of β-blocker, little real advantage is offered. Drugs with partial agonist activity may not increase peripheral resistance as much as those without.

Arterial pressure reduction begins within an hour of β-blocker administration, but several days may elapse before the plateau is reached and the full hypotensive effect of oral β-blockers takes about 2 weeks. This suggests the involvement of several mechanisms including readjustment of central and peripheral cardiovascular reflexes. During chronic administration, the hypotensive effects of β-blockers last longer than the pharmacological half-life, so that single daily dosage is adequate therapeutically. However, upregulation of receptors may occur, leading to adverse effects (tachycardia, hypertension, myocardial ischaemia) on abrupt withdrawal of β-blockers. This is important in surgical patients, and patients receiving long-term β-blockade should continue therapy throughout the perioperative period. There is some evidence that perioperative β-blockade reduces cardiac complications in high-risk patients undergoing major vascular surgery, but the data are conflicting. All β-blockers are equally effective as hypotensive drugs; patients unresponsive to one β-blocker are generally unresponsive to all.

Ischaemic heart disease. β-Blockers improve symptoms and decrease the frequency and severity of silent myocardial ischaemia in patients with ischaemic heart disease. The incidence of myocardial ischaemia in high-risk patients is reduced by perioperative β-blockade and long-term outcome may be improved. β-Blockers reduce heart rate and contractility, with consequent decreases in wall tension and myocardial oxygen demand. A slower heart rate also permits longer diastolic filling time and hence potentially greater coronary perfusion. The perfusion of ischaemic regions may be improved by redistribution of myocardial blood flow, and other additional mechanisms may be involved.

β-Blockade also reduces exercise-induced increases in arterial pressure, velocity of cardiac contraction and oxygen consumption at any workload. Partial agonists have less effect on the resting heart rate and theoretically increase the metabolic demand of the myocardium; they may be less effective in patients with angina at rest or at very low levels of exercise. In contrast to effects on arterial pressure, there is a more direct relationship between plasma concentration and antianginal effect. To achieve effective plasma concentrations over a sustained period as a single daily dosage, either the long half-life drugs (e.g. atenolol, nadolol) or slow-release preparations (e.g. oxprenolol-SR, propranolol-LA, metoprolol-SR) are required.

Secondary prevention of myocardial infarction. Early i.v. administration after acute myocardial infarction can decrease infarct size, the incidence of ventricular and supraventricular arrhythmias and mortality in both lower- and higher-risk groups (e.g. elderly patients or those with left ventricular dysfunction). Mortality is reduced by 20–40%, and the risk of re-infarction is reduced if oral therapy is continued for 2–3 years.

Obstructive cardiomyopathy. β-Blockers improve exercise tolerance and alleviate symptoms in hypertrophic obstructive cardiomyopathy by decreasing heart rate, myocardial work, contractility and, thus, outflow tract obstruction. However, the incidence of sudden death in this condition is not affected. The incidence of cyanotic episodes caused by pulmonary outflow obstruction in patients with Fallot’s tetralogy is reduced by a similar mechanism.

Congestive heart failure. Congestive heart failure is accompanied by a compensatory increase in sympathetic nervous stimulation with increased plasma and cardiac noradrenaline concentrations, leading to increases in cardiac output, systemic vascular resistance and afterload. Plasma renin concentration also increases. Although beneficial as an acute response in the short term, high circulating catecholamine concentrations are directly toxic to the myocardium. Desensitization of myocardial β1-adrenergic receptors occurs via downregulation and altered signal transduction. In combination with chronically increased peripheral resistance, this leads to ventricular remodelling with progressively worsening myocardial function and a propensity to arrhythmias. Some second- and third-generation β-blockers (bisoprolol, metoprolol and carvedilol) have been shown to decrease morbidity and mortality in heart failure by improving ventricular function. The mechanism is primarily by upregulation of β-receptor density or function, although other factors may contribute, including slowing of heart rate or an antiarrhythmic effect. Bisoprolol and metoprolol are β1-selective antagonists but carvedilol also has β2– and α1-antagonist and antioxidant effects which may contribute to its action. They must be introduced cautiously in heart failure because symptoms may initially worsen, and ventricular function improves only after 1 month of therapy.

Arrhythmias (see below). β-Blockers are effective in the treatment of arrhythmias caused by sympathetic nervous overactivity or after myocardial infarction. The mechanisms are related to β-blockade itself rather than any membrane-stabilizing effect, e.g. antagonism of catecholamine effects on the cardiac action potential and muscle contractility. The result is a slowing of rate of discharge from the sinus and any ectopic pacemaker, and slowing of conduction and increased refractoriness of the AV node. β-Blockers also slow conduction in anomalous pathways. They may be used i.v. to terminate an attack of supraventricular tachycardia or decrease the ventricular rate in atrial fibrillation and flutter; conversion to sinus rhythm may also be achieved. If given within 30 min of i.v. verapamil, there is a danger of severe bradycardia or asystole. Most β-blockers have similar antiarrhythmic effects in adequate dosage, but esmolol has the advantage of a short half-life (see below) so that adverse effects are limited. They are also useful as second-line alternatives for the treatment of ventricular tachycardia. Sotalol has both class 2 and class 3 antiarrhythmic activity (see below) and is licensed for use only for its antiarrhythmic action, in particular for the treatment of supraventricular and ventricular tachycardias.

Miscellaneous. β-Blockers are prescribed for migraine prophylaxis, essential tremor and anxiety states. They are useful for glaucoma because they decrease intraocular pressure, probably by reducing the production of aqueous humour. Topical preparations (e.g. timolol, betaxolol, carteolol) are used in an attempt to decrease adverse effects but significant systemic absorption may still take place; bradycardia, hypotension and bronchospasm may occur, particularly during anaesthesia. β-Blockers diminish the symptoms of thyrotoxicosis and are used as part of preoperative preparation before thyroidectomy. They may be used also as part of a hypotensive anaesthetic technique.

Reactions Resulting from β-Blockade:

Newer β-Blockers: Recently introduced third- generation β-blockers (e.g. labetalol, celiprolol, nevibolol and carvedilol) are mostly non-selective (β1 > β2) antagonists which also produce vasodilatation by several mechanisms. Labetalol and carvedilol are also α1-antagonists; bucindolol produces direct vasodilatation by a cAMP-dependent mechanism. The severity of some of the adverse effects of β-blockade may be less with those drugs with vasodilating properties.

Labetalol is a competitive α1-, β1– and β2-antagonist, which is a partial agonist at β2-receptors. It is four to seven times more potent at β- than at α-receptors and is useful for the prevention and treatment of perioperative hypertension, or to produce controlled hypotension (see Ch 21). It is also available as an oral preparation for the treatment of chronic hypertension or the preoperative management of phaeochromocytoma (see Ch 37). Intravenous labetalol in small increments (e.g. 5–10 mg) produces a controlled decrease in arterial pressure over 5–10 min with no change in cardiac output or reflex tachycardia, suggesting that at this dose the vasodilating action predominates. At higher doses, the β-effect becomes more prominent, with negative inotropic and chronotropic effects.

Carvedilol is an antagonist at α1– and β-receptors (with relative β:α1 selectivity of 10:1 and no partial agonist activity), but it has other effects including significant antioxidant activity, inhibition of endothelin synthesis and possibly calcium channel blockade in higher doses; these may account for some of its beneficial activity in patients with heart failure. Carvedilol is a stereoisomer which undergoes extensive first-pass metabolism with the production of active metabolites.

Nebivolol is a lipophilic β1-selective blocker which is administered as a racemic mixture of equal proportions of D– and L-enantiomers. It has no membrane-stabilizing activity but has vasodilatory effects probably mediated by endothelial nitric oxide.

Celiprolol is a β1-selective blocker which is a weak β2-agonist and has α2-antagonist activity. It also has direct vasodilator effects which may be mediated by endothelial nitric oxide release. Celiprolol is excreted unchanged.

Esmolol is a rapid-onset, short-acting β1-selective blocker with no membrane-stabilizing or partial agonist activity and is only available for i.v. use. It has an onset time of 1–2 min and is metabolized rapidly by red cell esterases (distinct from plasma cholinesterases); its elimination half-life is 9 min. The rapid onset and offset of effect are an advantage in the perioperative period because any effects such as bradycardia or hypotension are short-lived. It is effective in preventing or controlling intraoperative tachycardia and hypertension, and is also useful for the treatment of supraventricular tachyarrhythmias, e.g. atrial fibrillation or flutter. It may be given as a slow i.v. bolus of 0.5–2.0 mg kg–1 or an infusion of 25–500 μg kg–1 min–1; its effects terminate within 10–20 min of stopping the infusion.

DRUGS ACTING ON THE PARASYMPATHETIC NERVOUS SYSTEM

The major drugs in use which act on the parasympathetic nervous system are muscarinic antagonists (e.g. atropine, hyoscine and propantheline), and parasympathetic agonists (e.g. the anticholinesterases neostigmine and pyridostigmine). Neuromuscular blocking drugs act at nicotinic receptors, and are described in Chapter 6.

Parasympathetic Antagonists

Parasympathetic antagonists block muscarinic ACh receptors and are either tertiary or quaternary amine compounds. Tertiary amines, e.g. atropine and hyoscine, are more lipid-soluble and cross biological membranes, e.g. the blood–brain barrier, to affect central ACh receptors and produce sedative or stimulatory effects. Similar antimuscarinic drugs, e.g. orphenadrine, procyclidine, are useful in drug-induced Parkinson’s disease because of their predominant central action; procyclidine is also used for the reversal of acute dystonic reactions to dopaminergic drugs (e.g. phenothiazines, droperidol). Other muscarinic antagonists used as gastrointestinal or urinary antispasmodics are quaternary amines; they are poorly absorbed after oral administration and produce minimal central effects.

Atropine

Atropine has widespread, dose-dependent antimuscarinic effects on parasympathetic functions. Salivary secretion, micturition, bradycardia and visual accommodation are impaired sequentially. CNS effects (sedation or excitation, hallucinations and hyperthermia) may occur at high doses. Atropine is administered in doses of 0.6–3.0 mg i.v. to counteract bradycardia in the presence of hypotension and to prevent the bradycardia associated with vagal stimulation or the use of anticholinesterase drugs. Adverse cardiac effects of atropine include an increase in cardiac work and ventricular arrhythmias. Occasionally, atropine may produce an initial transient bradycardia, thought to be caused by increased ACh release mediated by M2-receptor antagonism. In therapeutic dosage, effects mediated by M3-receptors (tachycardia, bronchodilation, dry mouth, mydriasis) predominate.

VASODILATORS

Vasodilators dilate arteries or veins and may reduce afterload, preload or both. Acute and chronic heart failure are both associated with a reflex increase in sympathetic tone and an increase in systemic vascular resistance. By lowering this resistance (afterload), myocardial work and oxygen requirements are reduced. Vasodilators acting on the venous side of the circulation (e.g. nitrates) increase venous capacitance, reduce venous return to the heart and so decrease left ventricular filling pressure (preload), myocardial fibre length and myocardial oxygen consumption for the same degree of cardiac work performed. They have several clinical indications (Table 8.7).

TABLE 8.7

Indications for Vasodilators

Acute and chronic left ventricular failure

Prophylaxis and treatment of unstable and stable angina

Treatment of acute myocardial ischaemia and infarction

Chronic hypertension

Acute hypertensive episodes

Elective controlled hypotensive anaesthesia

Vasodilators may be classified into those acting directly on vascular smooth muscle (nitroprusside, nitrates, hydralazine, diazoxide, minoxidil, calcium channel blockers) and neurohumoral antagonists (α-blockers and ACE inhibitors). They may also be classified according to which side of the heart they act on preferentially. Hydralazine, calcium channel blockers, and minoxidil act mainly on afterload. Nitrates principally affect preload. Nitroprusside, α-blockers and ACE inhibitors have a balanced effect on arteries and veins.

Nitrates

The organic nitrates (glyceryl trinitrate and isosorbide mononitrate and dinitrate) cause systemic and coronary vasodilatation. They act primarily on systemic veins, causing venodilatation, sequestration of blood in venous capacitance beds and a reduction in preload. Arteriolar dilatation occurs at higher doses and afterload is reduced; tachycardia, hypotension and headaches may occur. Systolic pressure decreases more than diastolic pressure, so coronary perfusion pressure is preserved. In left ventricular failure, venodilatation is beneficial, reducing pulmonary congestion; cardiac dynamics may be improved so that stroke volume and cardiac output increase. Nitrates are used widely for the prevention and treatment of angina and myocardial infarction because they cause vasodilatation in stenotic coronary arteries and redistribution of myocardial blood flow. Glyceryl trinitrate (GTN) is a powerful myometrial relaxant. Nitrates also inhibit platelet aggregation in vitro.

Nitrates are converted to the active compounds nitric oxide (NO) and nitrosothiols by a denitration mechanism involving reduced sulphydryl groups. NO and nitrosothiols activate guanylate cyclase in the cytoplasm of vascular smooth muscle cells to increase intracellular cGMP. This leads to protein kinase phosphorylation and decreased intracellular calcium, causing vascular smooth muscle relaxation and vasodilatation. Tolerance to nitrates develops rapidly during continuous therapy (within 24 h), caused by depletion of reduced sulphydryl groups or activation of neurohormonal countermechanisms, and a daily nitrate-free interval of 8–12 h is required. Nitrates may be administered by oral, buccal, transdermal and intravenous routes. Intravenous nitrates may be used for the treatment of perioperative hypertension or myocardial ischaemia or as part of a deliberate hypotensive anaesthetic technique. They are absorbed by rubber and plastics (especially PVC infusion bags), so are best administered by syringe pump.

Sodium Nitroprusside

Sodium nitroprusside (SNP) is reduced to NO on exposure to reducing agents and in tissues, including vascular smooth muscle cell membranes. The process is non-enzymatic but SNP has a similar ultimate mechanism of action to nitrates (increased intracellular cGMP). SNP produces similar effects on capacitance and resistance vessels so that preload and afterload are equally reduced, and it is useful in the management of acute left ventricular failure. Systolic and diastolic pressures decrease equally in a dose-dependent manner. In larger doses (as used for hypotensive anaesthesia), heart rate increases.

Release of NO from nitroprusside is accompanied by release of cyanide ions, which are detoxified by the liver and kidney to thiocyanate (requiring thiosulphate, vitamin B12 and the enzyme rhodanase), which is excreted slowly in urine. It has an immediate, short-lived effect (lasting only for a few minutes) so it must be given by intravenous infusion. SNP is photodegraded to cyanide ions, so that infusion solutions should be protected from light and not used if they have turned dark brown or blue. Also, if the total dose of SNP exceeds 1.5 mg kg–1 or the infusion rate exceeds 1.5 μg kg–1 min–1, cyanide and thiocyanate may accumulate, with the risk of metabolic acidosis; plasma bicarbonate concentration should be monitored. The risks of cyanide toxicity are increased in the presence of impaired renal or hepatic function, and symptoms may be delayed until after the SNP infusion has been discontinued. Plasma cyanide or thiocyanate concentrations should also be monitored if the drug is used for more than 2 days. Thiocyanate is potentially neurotoxic and may cause hypothyroidism. In cases of suspected cyanide toxicity, sodium thiosulphate (which promotes conversion to thiocyanate), dicobalt edetate (which chelates cyanide ions) and hydroxocobalamin (which combines with cyanide to form cyanocobalamin) may be given. In practice, nitroprusside is usually well tolerated and most symptoms are associated with too rapid a decrease in arterial pressure.

Potassium Channel Activators

Hydralazine, minoxidil and diazoxide are direct-acting arteriolar vasodilators which have largely been superseded. Minoxidil and diazoxide activate ATP-sensitive K+ channels in vascular smooth muscle cells, causing K+ efflux and membrane hyperpolarization. This leads to closure of calcium channels, reduced intracellular calcium availability and consequently smooth muscle relaxation and arterial vasodilatation. Hydralazine may act via a similar mechanism. All these drugs reduce afterload, with little or no effect on preload. Their effects are limited by reflex tachycardia and a tendency to cause sodium and water retention (by activation of the renin-angiotensin system and a direct renal mechanism). Consequently, they are usually administered during long-term therapy with a β-blocker and a diuretic.

Hydralazine is the most widely used of these drugs. Its half-life is short (approximately 2.5 h) but its antihypertensive effect is relatively prolonged. It may be given as a slow i.v. bolus of 5–10 mg, with appropriate monitoring, for the treatment of hypertensive emergencies.

Minoxidil is only available orally. It has a long duration of action (12–24 h) unrelated to its plasma half-life, and it causes hypertrichosis. T-wave abnormalities on ECG are observed in 60% of patients.

Diazoxide has a similar structure to thiazide diuretics. It is occasionally used for the treatment of hypertensive emergencies; 1–3 mg kg–1 i.v. may be given rapidly (over 30 s) for effects lasting 4–24 h. However, it is difficult to control the action or duration of action of repeated doses.

CALCIUM CHANNEL BLOCKERS

Mechanism of Action

The normal function of cardiac myocytes and conducting tissues, skeletal muscle, vascular and other smooth muscle, and neurones depends on the availability of intracellular calcium ions. Under physiological conditions, calcium entry into the cell induces further calcium release from the sarcoplasmic reticulum, which facilitates conduction of the cardiac action potential and excitation–contraction coupling by interaction with calmodulin (in smooth muscle) or troponin (within cardiac muscle). Calcium enters the cell via several ion channels situated on the plasma membrane, the most important being voltage-gated calcium channels which are activated by nerve impulses or membrane depolarization. Other types of calcium channels are receptor-operated and stretch-activated channels. Calcium channel blockers (CCBs) are a diverse group of compounds which decrease calcium entry into cardiac and vascular smooth muscle cells through the L-subtype (long-lasting inward calcium current) of voltage-gated calcium channels. CCBs bind in several ways to the α1 subunit of L-type channels to impede calcium entry. Phenylalkylamines (e.g. verapamil) bind to the intracellular portion of the channel and physically occlude it, whereas dihydropyridines modify the extracellular allosteric structure of the channel. Benzothiazepines (e.g. diltiazem) act on the α1 subunit, although the mechanism has not been fully elucidated, and may have further actions on sodium-potassium exchange and calcium–calmodulin binding.

Cardiac cells in the SA and AV nodes depend on the slow inward calcium current for depolarization. CCBs which act here decrease calcium entry during phase 0 of the action potential of SA node and AV node cells, decreasing heart rate and AV node conduction. Calcium entry during phase 2 of the action potential of ventricular myocytes may be decreased (Fig. 8.5) and excitation–contraction coupling inhibited, causing decreased myocardial contractility. Some CCBs may also have favourable effects on endothelial function.

Clinical Effects

CCBs differ in their selectivity for cardiac muscle cells, conducting tissue and vascular smooth muscle, but they all decrease myocardial contractility and produce coronary and systemic vasodilatation with a consequent decrease in arterial pressure. They have been used widely for the treatment of hypertension and angina, but have been partly superseded by newer drugs. Other current indications include prevention of vasospasm in subarachnoid haemorrhage or Raynaud’s disease. Verapamil and diltiazem also decrease SA node activity, AV node conduction and heart rate, and they are useful in the treatment of paroxysmal supraventricular tachyarrhythmias. CCBs may also inhibit platelet aggregation, protect against bronchospasm and improve lower oesophageal sphincter function. The non-dihydropyridines are contraindicated in the presence of second- or third-degree heart block and should not be combined with β-blockers because they may cause bradycardia or heart block. With the exception of amlodipine and felodipine, calcium channel blockers should not be used in patients with heart failure. In some patients, sudden cessation of CCBs may lead to an exacerbation of angina symptoms.

Classification

CCBs are a diverse group of compounds which have been classified in several ways, according to their structure, mechanism of action and specificity for slow calcium channels. They are classified here by their chemical structure, tissue selectivity and pharmacokinetic properties (Table 8.8).

First-Generation Calcium Channel Blockers

The first-generation CCBs (verapamil, diltiazem and nifedipine) have a rapid onset of action which may reduce arterial pressure acutely and produce reflex sympathetic activation. They have marked negative dromotropic and inotropic effects (especially verapamil and diltiazem). Their intrinsic duration of action is short, but slow-release formulations have been developed (see below). All are well absorbed but undergo a significant first-pass effect, leading to low bioavailability. They are highly protein-bound and metabolized extensively by hepatic demethylation and dealkylation, with wide individual pharmacokinetic variability (Table 8.9). Most CCBs possess one or more chiral centres, and the different enantiomeric forms have different pharmacokinetic and pharmacodynamic properties. For example, L-verapamil undergoes higher first-pass metabolism than the D-form, so that plasma concentrations of L-verapamil are relatively higher after intravenous administration, producing more pronounced negative inotropic and chronotropic effects.

Nifedipine is a dihydropyridine derivative which is a systemic and coronary arterial vasodilator. It is effective in countering coronary artery spasm, thought to be an important component of all forms of angina, and it may bring symptomatic relief in patients with peripheral vasospastic (e.g. Raynaud’s) disease. Its antianginal effect is additive with that of β-adrenergic blocking drugs and nitrates. Adverse effects include flushing, headaches, ankle oedema, dizziness, tiredness and palpitations. Nifedipine is absorbed rapidly, particularly when the stomach is empty, with an onset of action of 20 min. This may produce reflex tachycardia and increased myocardial contractility.

Verapamil is a phenylalkylamine which has more pronounced effects on the SA and AV nodes compared with other CCBs, and is used mainly as an antiarrhythmic (see below). It has vasodilator and negative inotropic properties and is also used for the treatment of angina, hypertension and hypertrophic obstructive cardiomyopathy. Verapamil has a marked negative inotropic action and may cause bradycardia, hypotension, AV block or heart failure when combined with β-blockers or other cardiodepressant drugs (including volatile anaesthetic agents). It may also potentiate the effects of neuromuscular blocking drugs. Both verapamil and diltiazem inhibit the hepatic metabolism of several drugs; plasma concentrations of digoxin, carbamazepine and theophyllines are increased by verapamil.

Diltiazem is a benzothiazepine whose predominant effect is on coronary arteries rather than conducting tissue, and is used mainly in the treatment of hypertension and angina. It can cause myocardial depression, especially when combined with β-blockers. It is metabolized in the liver, producing active metabolites, and is excreted via the kidneys.

Second- and Third-Generation CCBs

The second-generation CCBs are dihydropyridine derivatives (either sustained-release formulations or new compounds) which have a slower onset and longer duration of action, and greater vascular smooth muscle selectivity. The slow onset results in less sympathetic activation and reflex tachycardia. The new compounds (e.g. felodipine, nisoldipine, nicardipine) have less effect on AV conduction and less negative inotropic and chronotropic effects. All have little effect on lipid or glucose metabolism and may be used in patients with renal dysfunction. Some have special features.

Nimodipine is selective for cerebral vasculature and is used to prevent vasospasm after subarachnoid haemorrhage. Nicardipine causes less reduction in myocardial contractility than other CCBs. Felodipine acts predominantly on peripheral vascular smooth muscle and has negligible effects on myocardial contractility, although it does produce coronary vasodilatation. It also has a mild diuretic and natriuretic effect. It is indicated for the treatment of hypertension, but has been used in patients with impaired LV function.

The third generation of CCBs (lacidipine, amlodipine) bind to specific high-affinity sites in the calcium channel complex. They have a particularly slow onset and long duration of action, and so reflex sympathetic stimulation is not evident but adverse effects related to vasodilatation (headache, flushing, ankle oedema) do occur. Both are extensively metabolized in the liver to inactive metabolites which are excreted via the kidneys and liver. Lacidipine is highly lipophilic, so that it is sequestered in the lipid bilayer of vascular smooth muscle cells and may delay the development of atherosclerosis via effects on modulators of vascular smooth muscle and platelet function. Lacidipine may augment the action of endothelium-derived relaxing factors (e.g. NO – which has vasodilator, antiplatelet and antiproliferative effects) and antagonize endothelin-1, a potent vasoconstrictor which also stimulates endothelial proliferation.

Anaesthesia and Calcium Channel Blockers

Both intravenous and volatile anaesthetic agents block conduction through L-type calcium channels in neuronal and cardiac tissues, and may therefore interact with CCBs through pharmacokinetic and pharmacodynamic mechanisms. In general, CCBs potentiate the hypotensive effects of volatile anaesthetics; verapamil (and to a lesser extent diltiazem) has additive effects with halothane on cardiac conduction and contractility, with the potential for bradycardia and myocardial depression. Verapamil decreases the MAC of halothane, and, in an animal model, nifedipine enhances the analgesic effects of morphine by stimulation of spinal 5-HT3 receptors. Plasma concentrations of verapamil are increased during anaesthesia with volatile agents, possibly because of decreased hepatic blood flow.

CCBs also potentiate the effects of depolarizing and non-depolarizing neuromuscular blockers in experimental conditions, although the clinical relevance of this is uncertain.

DRUGS ACTING VIA THE RENIN–ANGIOTENSIN–ALDOSTERONE SYSTEM

The renin–angiotensin–aldosterone system (RAS) is intimately involved with cardiovascular and body fluid homeostasis. Angiotensin II (AT-II) is the major regulator of the renin-angiotensin system and is a potent vasoconstrictor with several renal and extrarenal effects. AT-II has an important role in the maintenance of circulating volume in response to several stressors, while direct renal effects are mostly responsible for long-term regulation of body fluid volume and blood pressure.

The production of AT-II from angiotensinogen occurs in the walls of small blood vessels in the lungs, kidneys and other organs, and in the plasma. The rate-limiting step for this cascade is the plasma concentration of renin (Fig. 8.6). AT-II is metabolized by several peptidases to several breakdown products including angiotensin III (AT-III), which has some activity at angiotensin receptors. Four subtypes of angiotensin receptor have been defined (AT1–4). AT1-receptors are found principally in vascular smooth muscle, adrenal cortex, kidney, liver and some areas of the brain, and mediate all the known physiological functions of AT-II. AT2-receptors are present in the kidney, adrenal medulla, uterus, ovary and the brain; they may play a role in cell growth and differentiation. The roles of AT3– and AT4-receptors are unclear.

AT1-receptors are typical G-protein-coupled receptors which activate phospholipase C with the production of DAG and IP3. IP3 causes the release of intracellular Ca2 +, which activates enzymes to cause the phosphorylation of intracellular proteins. AT-II also increases Ca2 + entry through membrane channels.

AT-II (see Fig. 8.6) is a potent vasoconstrictor (by direct action on vascular smooth muscle of arterioles and veins) and it promotes sodium reabsorption both by direct action at the proximal tubules and by stimulating aldosterone secretion. AT-II produces preglomerular vasoconstriction and efferent arteriolar vasoconstriction, and so maintains glomerular filtration rate in response to a decrease in renal blood flow. It also affects the local regulation of blood flow in other vascular beds, e.g. the splanchnic circulation, and stimulates the sympathetic nervous system via direct and indirect methods to increase noradrenaline and adrenaline release; it may also inhibit cardiac vagal activity. AT-II stimulates erythropoiesis and has direct trophic effects on vascular smooth muscle and cardiac muscle, promoting cellular proliferation, migration and hypertrophy. There is some evidence for relative downregulation of AT1-receptors and upregulation of AT2-receptors in heart failure, with AT2-receptors being responsible for some cardioprotective effects.

RAS activity tends to be low in the resting state but renin production (the rate-limiting step in the production of AT-II) is activated by several stimuli, e.g. depletion of circulating volume, haemorrhage or sodium depletion. This increases AT-II production and causes vasoconstriction, increased sympathetic activity (with increased cardiac output and arterial pressure) and sodium retention. The RAS may be involved in the pathogenesis of hypertension but the relationship is complex; RAS activity may be high (e.g. in renal artery stenosis), low (as in primary aldosteronism) or variable (essential hypertension).

Drugs Acting on the Renin–Angiotensin System

The activity of the RAS may be inhibited by several mechanisms:

image Suppression of renin release or inhibition of renin activity. Sympatholytic drugs (e.g. β-blockers or central α-antagonists) directly inhibit renin secretion. Renin inhibitors competitively inhibit the reaction between renin and angiotensinogen, preventing the production of AT-II. Because renin release still occurs, the consequent reduction of plasma AT-II concentrations leads to a secondary increase in renin secretion, limiting the effect of such drugs.

image Inhibition of angiotensin-converting enzyme (ACE). The primary mechanism of action of ACE inhibitors is to block the conversion of angiotensin I (AT-I) to AT-II, although effects on kinin and prostaglandin metabolism also contribute.

image Blockade of ATII receptors. AT1-receptor blockers (ARBs) non-competitively block AT1-receptors and inhibit the RAS independently of the source of AT-II. They block any effects of AT-II resulting from compensatory stimulation of renin, such as reflex activation of the sympathetic nervous system.

image Aldosterone antagonism. Spironolactone and eplerenone are competitive antagonists of aldosterone at renal nuclear mineralocorticoid receptors (see Ch 10).

ACE Inhibitors

ACE inhibitors act principally by inhibition of AT-II formation, but effects on the kallikrein-kinin system are also important. All ACE inhibitors reduce arteriolar tone, peripheral resistance and arterial pressure by decreasing both AT-II-mediated vasoconstriction and sympathetic nervous system activity. Renal blood flow increases, further inhibiting aldosterone and antidiuretic hormone secretion and promoting sodium excretion. ACE inhibitors are useful in patients with heart failure because preload and afterload decrease without an increase in heart rate, and cardiac output increases.

ACE is the same enzyme as kininase II and is involved in the metabolism of both kinins and prostaglandins. ACE inhibitors therefore block the degradation of kinins, substance P and endorphins, and increase prostaglandin concentrations. Bradykinin and other kinins are highly potent arterial and venous dilators which stimulate the production of arachidonic acid metabolites, NO and endothelial-derived hyperpolarization factor via specific bradykinin β2-receptors in vascular endothelium. Bradykinin also enhances the uptake of circulating glucose into skeletal muscle and has a protective effect on cardiac myocytes by a mechanism involving prostacyclin stimulation. Kinins have no major effect on arterial pressure regulation in normotensive individuals or those with low-renin hypertension, but they account for up to 30% of the effects of ACE inhibitors in renovascular hypertension. The adverse effects of dry cough and angioneurotic oedema sometimes associated with ACE inhibitors may be kinin-dependent. ACE is widely distributed in tissues and plasma; ACE inhibitors may differ in their affinity for ACE at different sites. Other tissue enzymes (AT-I convertase and chymase) may also produce AT-II, from AT-I or directly from angiotensinogen, so that ACE inhibitors do not completely block RAS activity.

Clinical Applications of ACE Inhibitors: ACE inhibitors are established in the treatment of hypertension and congestive heart failure. They improve left ventricular dysfunction after myocardial infarction, delay the progression of diabetic nephropathy and have a protective effect in non-diabetic chronic renal failure. ACE inhibitors improve vascular endothelial function by their effects on AT-II and bradykinin, and improve long-term cardiovascular outcome in patients with established vascular disease. ACE inhibitors have a common mechanism of action, differing in the chemical structure of their active moieties in potency, bioavailability, plasma half-life, route of elimination, distribution and affinity for tissue-bound ACE (Table 8.10). Most of the newer compounds are prodrugs, converted to an active metabolite by the liver, and have a prolonged duration of action. Most are excreted via the kidneys, and dosage should be reduced in the elderly and those with impaired renal or cardiac function. Enalapril is also available as the active drug, enalaprilat, and may be administered i.v. for the treatment of hypertensive emergencies.

ACE inhibitors are generally well tolerated, with no rebound hypertension after stopping therapy and few metabolic effects. Symptomatic first-dose hypotension may occur, particularly in hypovolaemic or sodium-depleted patients with high plasma renin concentrations. Symptomatic hypotension was more common with the higher doses originally used. ACE inhibitors have a synergistic effect with diuretics (which increase the activity of the renin-angiotensin system) but are less effective in patients taking NSAIDs.

Adverse effects of ACE inhibitors are classified into those that are class-specific (related to inhibition of ACE) and those which relate to specific drugs. Class-specific effects include hypotension, renal insufficiency, hyperkalaemia, cough (10%) and angioneurotic oedema (0.1–0.2%). ACE inhibitors may cause renal impairment, particularly if renal perfusion is decreased (e.g. because of renal artery stenosis, congestive heart failure or hypovolaemia) or if there is pre-existing renal disease. Renal impairment is also more likely in the elderly or those receiving NSAIDs, and renal function should be checked before starting ACE inhibitor therapy, and monitored subsequently. Hyperkalaemia (plasma K+ concentration usually increases by 0.1–0.2 mmol L–1 because of decreased aldosterone concentrations) may be more marked in those with impaired renal function or in patients taking potassium supplements or potassium-sparing diuretics. The mechanism of cough is not known but is mediated by C fibres and may be related to bradykinin or substance P production. It is reversible on stopping the ACE inhibitor. Other adverse effects include upper respiratory congestion, rhinorrhoea, gastrointestinal disturbances, and increased insulin sensitivity and hyperglycaemia in diabetic patients.

Some adverse effects, e.g. skin rashes (1%), taste disturbances, proteinuria (1%) and neutropenia (0.05%), are related to the presence of a sulphydryl group (e.g. captopril). ACE inhibitors are contraindicated in pregnancy.

Although anaesthesia per se has no direct effect on the RAS or ACE inhibitors, the RAS is activated by several stimuli which may occur during the perioperative period. These include blood or fluid losses and the stress response to surgical stimulation. RAS activation contributes to the maintenance of arterial pressure after haemorrhage, or during anaesthesia. Refractory hypotension during anaesthesia has been reported in patients receiving long-term antihypertensive treatment with ACE inhibitors, and it has been recommended that they are stopped 24 h before surgery if significant blood loss or fluid shifts are likely. ACE inhibitors improve ventricular function in patients with heart failure or after myocardial infarction but it is not known whether acute cessation before surgery is harmful. Conversely, they may have beneficial effects on regional blood flow and have been associated with improved renal function in patients undergoing aortic surgery.

Angiotensin-II Receptor Blockers

Angiotensin-II receptor blockers (ARBs) specifically and non-competitively block the AT1-receptor, inhibiting the RAS independently and blocking any effects of AT-II resulting from compensatory stimulation of renin. Hence, they reduce afterload and increase cardiac output without causing tachycardia and are used in the treatment of hypertension, diabetic nephropathy and heart failure. As they have no effect on bradykinin metabolism or prostaglandin synthesis, ARBs do not produce the cough or rash associated with ACE inhibitors, though angio-oedema has been reported. However, plasma renin, AT-I and AT-II concentrations increase, and aldosterone concentrations decrease, during long-term therapy: hyperkalaemia may occur if potassium-sparing diuretics are also administered. Losartan is also uricosuric. All available ARBs are non-peptide imidazole compounds and are highly protein bound, with a prolonged duration of action exceeding their plasma half-life, and a maximum antihypertensive effect 2–4 weeks after starting therapy. In common with ACE inhibitors, they are contraindicated in pregnancy and are likely to have an adverse effect in patients with renal artery stenosis or those taking NSAIDs. The pharmacological properties of some ARBs are shown in Table 8.11. There are few data describing the effects of ARBs in the perioperative period, but caution would be appropriate when large fluid or blood losses are expected.

ANTIARRHYTHMIC DRUGS

Cardiac arrhythmias are irregular or abnormal heart rhythms and include bradycardias or tachycardias outside the physiological range. Patients may present for surgery with a pre-existing arrhythmia; alternatively, arrhythmias may be precipitated or accentuated during anaesthesia by several surgical, pharmacological or physiological factors (Table 8.12). Although several drugs (including anaesthetic drugs) have effects on heart rate and rhythm, the term antiarrhythmic is applied to drugs which primarily affect ionic currents within myocardial conducting tissue. Therapy for long-term arrhythmias has changed during the last two decades with the development of non- pharmacological techniques, e.g. DC cardioversion, implantable cardioverter-defibrillator (ICD) devices or radiofrequency ablation of ectopic foci. Most forms of supraventricular tachycardia may be controlled by radiofrequency ablation and ICDs are increasingly used in patients who have suffered an episode of ventricular tachycardia. Long-term drug therapy has therefore declined and is now largely confined to patients with atrial fibrillation, or as an adjunct in patients with ICDs or benign arrhythmias resistant to catheter ablation. However, owing to the frequency of arrhythmias during anaesthesia, knowledge of the available drugs and their interactions is important for the anaesthetist. Antiarrhythmic drugs are classified according to their effects on the action potential (see below).

TABLE 8.12

Precipitants of Arrhythmias During Anaesthesia

Myocardial ischaemia

Hypoxia

Hypercapnia

Halogenated hydrocarbons (volatile anaesthetic agents, e.g. trichloroethylene, cyclopropane, halothane)

Catecholamines (endogenous or exogenous)

Electrolyte abnormalities (hypo- or hyperkalaemia, hypocalcaemia, hypomagnesaemia)

Hypotension

Autonomic effects (e.g. reflex vagal stimulation, brain tumours or trauma)

Acid–base abnormalities

Mechanical stimuli (e.g. during CVP or PAFC catheter insertion)

Drugs (toxicity or adverse reactions)

Medical conditions (e.g. sepsis, myocarditis, pneumonia, alcohol abuse, thyrotoxicosis)

The Cardiac Action Potential

The cardiac action potential (AP) is generated by movement of charged ions across the cell membrane and comprises five phases (see Fig. 8.5). At rest, the cells are polarized and the resting membrane potential is negative (− 50 to − 60 mV in sinus node pacemaker cells and − 80 to − 90 mV in Purkinje, atrial and ventricular muscle fibres). The AP is triggered by a low intra-cellular leak of Na+ ions (and Ca2 + ions at the AV node) until a threshold point is reached, when sudden rapid influx of Na+ ions causes an increase in positive charge within the cell and generates an impulse (phase 0, depolarization). The AP starts to reverse (phase 1), but is sustained because of slower inward movement of Ca2 + ions (phase 2). Efflux of K+ ions brings about repolarization (phase 3) and the gradual termination of the AP. Thereafter, re-equilibration of Na+ and K+ takes place and the resting membrane potential is restored (phase 4). The AP spreads between adjacent cells and is transmitted through the specialized conducting system from the AV node to the bundle of His and ventricular muscle fibres via the Purkinje fibres. The SA node pacemaker cells have the fastest spontaneous discharge rate and usually initiate the coordinated action potential. However, action potentials may also be generated by the AV node and other cells in the conducting system.

Mechanisms of Arrhythmias

Arrhythmias are caused by abnormalities of impulse generation or conduction, or both, via a number of mechanisms:

image Altered automaticity. Increased pacemaker activity in the SA node (e.g. caused by increased sympathetic tone) may cause sinus tachycardia, or atrial or ventricular tachyarrhythmias. Decreased SA node automaticity (e.g. as a result of enhanced vagal activity) may allow the emergence of latent pacemaker activity in distal conducting tissues, e.g. AV node or the bundle of His–Purkinje system, causing sinus bradycardia, AV nodal or idioventricular escape rhythms. These rhythms are common during halothane anaesthesia.

image Unidirectional conduction block. Interruption of the normal conduction pathways caused by anatomical defects, alterations in refractory period or excitability may cause heart block and favours arrhythmias caused by abnormal re-entry or automaticity.

image Ectopic foci. Ectopic foci may give rise to arrhythmias in a variety of circumstances. In the presence of bradycardia or SA node block, pathological damage in cardiac muscle cells or conducting tissues may augment the generation of arrhythmias from ectopic foci, via:

Other factors may contribute. Increased automaticity in atrial, ventricular or conducting tissues caused by ischaemia or electrolyte disturbances (e.g. hypokalaemia) may trigger depolarization before the SA node and cause an arrhythmia. Re-entrant arrhythmias arise when forward conduction of impulses in a branch of the conduction pathway is blocked by disease and retrograde conduction occurs. If there is a discrepancy in the refractory periods of the two branches, retrograde conduction may occur in cells which have already discharged and repolarized, triggering a further AP which is both premature and ectopic. These premature APs become self-sustaining (circus movements), leading to atrial or ventricular tachycardia or fibrillation. Pathological afterdepolarizations are spontaneous impulses arising just after the normal AP and occur mostly in ischaemic myocardium (e.g. after myocardial infarction), especially in the presence of hypoxaemia, increased catecholamine concentrations, digoxin toxicity or electrolyte abnormalities.

Arrhythmias and Anaesthesia

Arrhythmias are common during anaesthesia and in intensive care, especially in patients with preoperative arrhythmia, cardiomyopathy, ischaemia or valvular or pericardial disease. They may be precipitated by several factors (see Table 8.13). Some arrhythmias are immediately life-threatening but all warrant attention because they usually imply the presence of other disturbances, and the effects of specific therapy (e.g. drugs, electrical cardioversion or cardiac pacing) are enhanced by prior corrective measures. Specific antiarrhythmic treatment is usually reserved for those arrhythmias affecting cardiac output or those which may progress to dangerous tachyarrhythmias.

In addition to volatile agents, several drugs used during anaesthesia may facilitate arrhythmias by direct toxicity (e.g. local anaesthetics), autonomic effects (e.g. succinylcholine, pancuronium), by enhancing the effects of catecholamines (e.g. nitrous oxide, thiopental, cocaine) or as a result of histamine release. Opioids potentiate central vagal activity, decrease sympathetic tone and have direct negative chronotropic effects on the SA node. They may therefore cause bradycardia but decrease the incidence of ventricular arrhythmias.

Mechanisms of Action of Antiarrhythmic Drugs

An arrhythmia may be controlled either by slowing the primary mechanism or, in the case of supraventricular arrhythmias, by reducing the proportion of impulses transmitted through the AV node to the ventricular conducting system. The cardiac action potential may be pharmacologically manipulated in three ways:

image The automaticity (tendency to spontaneous discharge) of cells may be reduced. This result can be achieved by reducing the rate of leakage of sodium (reducing the slope of phase 4), by increasing the electronegativity of the resulting membrane potential or by decreasing the electronegativity of the threshold potential.

image The speed of conduction of the action potential may be suppressed as reflected by a lowering of the height and slope of the phase 0 discharge. A reduction in the electronegativity of the membrane potential at the onset of phase 0 reduces both the amplitude and the slope of the phase 0 depolarization. This situation occurs if the cell discharges before it has been completely repolarized.

image The rate of repolarization may be reduced, which prolongs the refractory period of the discharging cell.

All antiarrhythmic drugs may themselves induce arrhythmias. Many (particularly class 1 antiarrhythmics) have a narrow therapeutic index and some have been associated with an increase in mortality in large-scale studies. Antiarrhythmic agents may be classified empirically on the basis of their effectiveness in supraventricular tachycardias (e.g. digoxin, β-blockers and verapamil) or in ventricular arrhythmias (lidocaine, mexiletine, magnesium, phenytoin). Many drugs (disopyramide, amiodarone, quinidine and procainamide) are effective in both supraventricular and ventricular arrhythmias (Table 8.13). The Vaughan Williams classification (Table 8.14) is based on electrophysiological mechanisms. This classification has limitations (some drugs belong to more than one class, some arrhythmias may be caused by several mechanisms, some drugs, e.g. digoxin and adenosine, do not fit into the classification) but it remains in use and is therefore described below.

Class 1 antiarrhythmic drugs (Table 8.15) inhibit the fast Na+ influx during depolarization; they inhibit arrhythmias caused by abnormal automaticity or re-entry. All class 1 drugs decrease the maximum rate of rise of phase 0, and decrease conduction velocity, excitability and automaticity to varying degrees. In addition to these local anaesthetic properties, some have membrane-stabilizing effects. Class 1a drugs antagonize primarily the fast influx of Na+ ions and so reduce conduction velocity through the AV node and His–Purkinje system, whilst prolonging the duration of the action potential and the refractory period. They also have varying antimuscarinic and sympathomimetic effects. Class 1b drugs have much less effect on conduction velocity in usual therapeutic doses and they shorten the refractory period. Agents in class 1c affect conduction profoundly without altering the refractory period.

β-Blockers (class 2) depress automaticity in the SA and AV nodes, and attenuate the effects of catecholamines on automaticity and conduction velocity in the sinus and AV nodes. Class 3 drugs prolong the AP and so lengthen the refractory period. Verapamil (class 4) also prolongs the AP, in addition to depressing automaticity (especially in the AV node).

Class 1 Antiarrhythmics

Class 1a: These drugs (see Tables 8.14 and 8.15) are used for the treatment and prevention of ventricular and supraventricular arrhythmias. Their use in the prevention of atrial fibrillation has declined because of proarrhythmic effects causing increased mortality, especially in patients with ischaemic heart disease or poor LV function. They may induce torsades de pointes (a form of polymorphic ventricular tachycardia) even in patients without structural heart disease.

Quinidine is an isomer of quinine with antimuscarinic and α-blocking properties. It was formerly used in the treatment of atrial and supraventricular tachycardias. Disopyramide is useful in supraventricular tachycardias and as a second-line agent to lidocaine in ventricular arrhythmias. It has less action on the His–Purkinje system than quinidine, but greater antimuscarinic and negative inotropic effects.

Procainamide has similar effects to quinidine and may cause hypotension after i.v. administration. It may be used i.v. to terminate ventricular arrhythmias or as an oral antiarrhythmic, although it has a short half-life and requires frequent administration or the use of a sustained-release oral preparation.

Ajmalin is a quinidine-like drug used for the treatment of Wolff–Parkinson–White (WPW) syndrome. It inhibits intraventricular conduction and prolongs AV conduction time, and is available in Europe.

Class 1b: Class 1b drugs are useful for the prevention and treatment of premature ventricular contractions, ventricular tachycardia and ventricular fibrillation, particularly associated with ischaemia.

Lidocaine is the first-choice drug for ventricular arrhythmias resistant to DC cardioversion. It decreases normal and abnormal automaticity and decreases action potential and refractory period durations. The threshold for ventricular fibrillation is raised but it has minimal haemodynamic effects. The antiarrhythmic properties of lidocaine are enhanced by hypoxaemia, acidosis and hyperkalaemia, so that it is particularly effective in ischaemic cells, e.g. after acute myocardial infarction, during cardiac surgery or in arrhythmias associated with digitalis toxicity. Lidocaine may cause CNS toxicity. Cardiotoxic effects (hypotension, bradycardia or heart block) occur at higher doses and are potentiated by hypoxaemia, acidosis and hypercapnia. Clearance is decreased if hepatic blood flow is decreased (e.g. in the elderly, those with congestive heart failure, after myocardial infarction), and also by β-blockers, cimetidine and liver disease. In these circumstances, the dose should be reduced by 50%. It is less effective in the presence of hypokalaemia.

Mexiletine is a lidocaine analogue which is well absorbed orally with high bioavailability. Adverse effects are similar to those of lidocaine; nausea and vomiting are also common during oral treatment.

Class 1c: Class 1c drugs are used for the prevention and treatment of supraventricular and ventricular tachyarrhythmias and junctional tachycardias with or without an accessory pathway. They are proarrhythmogenic, particularly in patients with myocardial ischaemia, poor left ventricular function or after myocardial infarction, and although effective in chronic atrial fibrillation, they are reserved for life-threatening arrhythmias.

Flecainide is a procainamide derivative with little effect on repolarization, the refractory period or AP duration, but unlike other drugs in this class, it decreases automaticity and produces dose-dependent widening of the QRS complex. In acute atrial fibrillation, intravenous flecainide usually restores sinus rhythm and is useful prophylaxis against further episodes of atrial fibrillation. However, it increases the risk of ventricular arrhythmias after myocardial infarction, especially in patients with structural cardiac disease.

Propafenone has a complex pharmacology including weak antimuscarinic, β-adrenergic receptor and calcium channel blocking effects. It should be used with caution in patients with reactive airways disease. Interaction with digoxin may increase plasma digoxin concentrations.

Class 3 Antiarrhythmic Drugs

Class 3 antiarrhythmics prolong the AP in conducting tissues and myocardial muscle. They prolong repolarization by K+ channel blockade, and decrease outward K+ conduction in the bundle of His, atrial and ventricular muscle, and accessory pathways. They are used for the treatment of supraventricular and ventricular tachyarrhythmias, including those associated with accessory conduction pathways. Some drugs have other actions (e.g. sotalol also produces β-blockade, and disopyramide has class 1 effects). All may prolong the QT interval and precipitate torsades de pointes, especially in high doses or in the presence of electrolyte disturbance.

Sotalol is a non-selective β-blocker with class 3 antiarrhythmic effects. Action potential duration and refractory period are lengthened, and it is effective in the treatment of supraventricular tachyarrhythmias, especially atrial flutter and fibrillation, which may be converted to sinus rhythm. It also suppresses ventricular tachyarrhythmias and ventricular ectopic beats. However, sotalol may cause torsades de pointes and other life-threatening arrhythmias, particularly in the presence of hypokalaemia.

Amiodarone is primarily a class 3 drug; it acts by inhibition of inward K+ current. It also blocks sodium and calcium channels, and has competitive inhibitory actions at α- and β-adrenoceptors, and may therefore be considered to have class 1, 2 and 4 antiarrhythmic activity. It prolongs AP duration, repolarization and refractory periods in the atria and ventricles. In addition, AV node conduction is markedly slowed and refractory period increased. Ventricular conduction velocity is slowed. Amiodarone is effective against a wide variety of supraventricular and ventricular arrhythmias, including WPW syndrome, and is preferred to other drugs in the presence of left ventricular dysfunction. Intravenous amiodarone is contraindicated in the presence of bradycardia or AV block but is less likely than other agents to cause arrhythmias. Bradycardia unresponsive to atropine, and hypotension, have been reported during general anaesthesia in patients receiving amiodarone therapy. Long-term oral therapy may produce a number of adverse effects. Amiodarone is an iodinated compound, which explains its effects on the thyroid (Table 8.16).

Bretylium is a quaternary ammonium compound which prevents noradrenaline uptake into sympathetic nerve endings. It prolongs AP duration and refractory period with no effect on automaticity, and was used as second-line therapy to lidocaine for resistant life-threatening ventricular tachyarrhythmias, but is no longer available in the UK.

Dronaderone and dofetilide are new class 3 antiarrhythmics which selectively inhibit inward K+ currents and so prolong repolarization, effective refractory period and the QT interval. Dofetilide has high bioavailability and is excreted mostly unchanged by the kidney but may cause torsades de pointes. Dronaderone is an analogue of amiodarone which also blocks multiple channels including L-type Ca2 + and inward Na+ currents. Both are used for the long-term treatment of atrial fibrillation.

Class 4 Antiarrhythmic Drugs

Class 4 drugs (calcium channel blockers) prevent voltage-dependent calcium influx during depolarization, particularly in the SA and AV nodes. Verapamil is more selective for cardiac cells than other calcium channel blockers but it is also a coronary and peripheral vasodilator, and decreases myocardial contractility. It depresses AV conduction, is effective in supraventricular or re-entrant tachycardia and controls the ventricular rate in atrial fibrillation. However, it is contraindicated in WPW syndrome because conduction through the accessory pathway may be encouraged, leading to ventricular fibrillation. Intravenous administration may cause hypotension (by vasodilatation), and caution is necessary in low-output states and in patients treated with negative inotropic drugs, e.g. β-blockers, disopyramide, quinidine or procainamide. Verapamil and diltiazem are effective by both i.v. and oral routes.

Other Antiarrhythmics

Adenosine is an endogenous purine nucleoside which mediates a variety of natural cellular functions via membrane-bound adenosine receptors, of which several subtypes (A1–A4) have been identified. Myocardial A1-receptors activate potassium channels and decrease cAMP by activating inhibitory Gi-proteins; A2-receptors mediate coronary vasodilatation by stimulating endothelial-derived relaxing factor and increasing intracellular cAMP. Increased potassium conductance induces membrane hyperpolarization in the SA and AV nodes, reducing automaticity and blocking AV node conduction. Adenosine also has an antiadrenergic effect in calcium-dependent ventricular tissue. It effectively converts paroxysmal supraventricular tachyarrhythmias (including those associated with WPW syndrome) to sinus rhythm, is used in the diagnosis of broad complex tachycardias when the origin (ventricular or supraventricular) is uncertain but is ineffective in the conversion of atrial flutter or fibrillation. In patients unable to exercise, adenosine is used as a coronary vasodilator in combination with myocardial perfusion scanning to diagnose coronary artery disease. Adenosine is metabolized to AMP or inosine by erythrocytes and vascular endothelial cells, so that it has a very short duration of action and is given as a rapid i.v. bolus.

Magnesium sulphate. Magnesium is a cofactor for many enzyme systems, including myocardial Na+/K+ ATPase. It antagonizes atrial L and T type Ca2 + channels, so that it prolongs both atrial refractory periods and conduction, and also inhibits K+ entry and suppresses ventricular afterdepolarizations. Intravenous magnesium sulphate is the treatment of choice for torsades de pointes, a type of ventricular tachycardia occasionally induced by class 1a or class 3 antiarrhythmic drugs which prolong the QT interval. It is a second-line treatment for supraventricular and ventricular arrhythmias, particularly those associated with digoxin toxicity or hypokalaemia, and is used as an anticonvulsant in patients with pre-eclampsia. Magnesium is redistributed rapidly into bone (50%) and intracellular fluid (45%), with the remainder excreted via the kidneys. It is therefore administered as an i.v. infusion.

Cardiac Glycosides

Digoxin and digitoxin are cardiac glycosides derived from plant sources, principally Digitalis purpura and Digitalis lanata. Their structure comprises a cyclopentanophenanthrene nucleus, an aglycone ring (responsible for the pharmacological activity) and a carbohydrate chain made up of sugar molecules (which aid solubility). Digitalis compounds have been used for over 200 years for the treatment of heart failure but have been largely superseded and are now principally indicated for the control of ventricular rate in supraventricular arrhythmias, particularly atrial fibrillation. They have several actions, including direct effects on the myocardium and both direct and indirect actions on the ANS. They increase myocardial contractility and decrease conduction in the AV node and bundle of His. Action potential and refractory period durations in atrial cells are reduced, and the rate of phase 4 depolarization in the SA node (automaticity) is decreased. The refractory periods of the AV node and bundle of His are increased, but in the ventricles, refractory period is decreased and spontaneous depolarization rate increases. This increased ventricular excitability is more marked in the presence of hypokalaemia and may lead to the appearance of ectopic pacemaker foci. The principal direct cardiac action is inhibition of membrane Na+/K+-ATPase activity. Intracellular Na+ concentration and Na+/Ca2 + exchange increase, leading to increased availability of intracellular Ca2 + and increased myocardial contractility. Increased local catecholamine concentrations as a result of decreased neuronal re-uptake and increased central sympathetic drive may also contribute to this positive inotropic action. In addition to direct cardiac actions, digitalis compounds have direct and indirect vagal effects. Central vagal tone, cardiac sensitivity to vagal stimulation, and local myocardial concentrations of acetylcholine are all increased and these effects may be partly antagonized by atropine.

Digoxin has a large apparent volume of distribution and a long half-life, so that effective plasma concentrations occur after approximately 5–7 days unless a loading dose is given. Doses should be reduced in renal impairment or elderly patients. The therapeutic index is low and toxicity is likely at plasma concentrations > 2.5 ng mL–1. However, plasma concentrations are a poor guide to toxicity because the drug is concentrated in cardiac and other tissues. Even at therapeutic plasma concentrations, digitalis affects the ECG, causing repolarization abnormalities. The classic ‘digoxin effect’ on ECG is of downsloping (‘reverse tick’) ST-segment depression with T wave inversion which may be wrongly interpreted as ischaemia. These changes are usually widespread, are not confined to the territory of one coronary artery and do not indicate toxicity. Digoxin toxicity usually causes cardiac, CNS, visual and gastrointestinal disturbances, including almost any arrhythmia, although ventricular arrhythmias (extrasystoles, bigeminy and trigeminy) and various degrees of heart block are commonest. Supraventricular arrhythmias also occur, often with some degree of conduction block. Digoxin should be avoided in the presence of second-degree heart block, ventricular tachycardia or aberrant conduction pathways (e.g. WPW syndrome) because arrhythmias may be precipitated, and should be used with caution after myocardial infarction. Sensitivity to digoxin is increased by hypokalaemia, hypomagnesaemia, hypercalcaemia, renal impairment, chronic pulmonary or heart disease, myxoedema and hypoxaemia. β-Blockers and verapamil have combined effects on the AV node and digoxin should be administered cautiously. Treatment of serious arrhythmias involves careful administration of KCl under ECG monitoring (especially in the presence of heart block or renal impairment). Lidocaine and phenytoin are useful for ventricular arrhythmias, β-blockade for supraventricular arrhythmias, and bradyarrhythmias may be treated with atropine. Digoxin should be stopped for at least 48 h before elective DC cardioversion, otherwise ventricular fibrillation may be precipitated. If cardioversion is required, the initial energy level should be low (e.g. 10–25 J) and increased if necessary.

Digitoxin is metabolized by the liver, and is less dependent upon renal function for its elimination. It has a very long half-life (4–6 days), so maintenance doses may be required only on alternate days, but this is also a disadvantage as toxic effects are very persistent.