Basic & clinical pharmacology 2

Page 1

These NOS isoforms generate NO from the amino acid L-arginine in an O2 - and NADPH-dependent reaction (Figure 19–1). This enzymatic reaction involves enzyme-bound cofactors, including heme, tetrahydrobiopterin, and flavin adenine dinucleotide (FAD). In the case of nNOS and eNOS, NO synthesis is triggered by agents and processes that increase cytosolic calcium concentrations. Cytosolic calcium forms complexes with calmodulin, an abundant calcium-binding protein, which then binds and activates eNOS and nNOS. On the other hand, iNOS is not regulated by calcium, but is constitutively active. In macrophages and several other cell types, inflammatory mediators induce the transcriptional activation of the iNOS gene, resulting in accumulation of iNOS and increased synthesis of NO.


FIGURE 19–1 Synthesis and reactions of nitric oxide (NO). L-NMMA (see Table 19-3) inhibits nitric oxide synthase. NO binds to the iron in hemoproteins (eg, guanylyl cyclase), resulting in the activation of cyclic guanosine monophosphate (cGMP) synthesis and cGMP target proteins such as protein kinase G. Under conditions of oxidative stress, NO can react with superoxide to nitrate tyrosine. GTP, guanosine triphosphate.

Signaling Mechanisms NO mediates its effects by covalent modification of proteins. There are three major targets of NO (Figure 19–1): 1. Metalloproteins—NO interacts with metals, especially iron in heme. The major target of NO is soluble guanylyl cyclase (sGC), a heme-containing enzyme that generates cyclic guanosine monophosphate (cGMP) from guanosine triphosphate (GTP). NO binds to the heme in sGC, resulting in enzyme activation and elevation in intracellular cGMP levels. cGMP activates protein kinase G (PKG), which phosphorylates specific proteins. In blood vessels, NO-dependent elevations in cGMP and PKG activity result in the phosphorylation of proteins that lead to reduced cytosolic calcium levels and subsequently reduced contraction of vascular smooth muscle. Interaction of NO with other metallo-proteins mediates some of the cytotoxic effects of NO associated with NO overproduction, eg, by activated macrophages. For example, NO inhibits metalloproteins involved in cellular respiration, such as the citric acid cycle enzyme aconitase and the electron transport chain protein cytochrome oxidase. Inhibition of heme-containing cytochrome P450 enzymes by NO is a major pathogenic mechanism in inflammatory liver disease. 2. Thiols—NO reacts with thiols (compounds containing the –SH group) to form nitrosothiols. In proteins, the thiol moiety is found in the amino acid cysteine. This posttranslational modification, termed S-nitrosylation or S-nitrosation, requires either metals or O2 to catalyze the formation of the nitrosothiol adduct. S-nitrosylation is highly specific, with only certain cysteine residues in proteins becoming Snitrosylated. S-nitrosylation can alter the function, stability, or localization of target proteins. Major targets of S-nitrosylation include H-


ras, a regulator of cell proliferation that is activated by S-nitrosylation, and the metabolic enzyme glyceraldehyde-3-phosphate dehydrogenase, which is inhibited when it is S-nitrosylated. Denitrosylation of proteins is poorly understood but may involve enzymes, such as thioredoxin, or chemical reduction by intracellular reducing agents such as glutathione, an abundant intracellular sulfhydrylcontaining compound. Glutathione can also be S-nitrosylated under physiologic conditions to generate S-nitrosoglutathione. Snitrosoglutathione may serve as an endogenous stabilized form of NO or as a carrier of NO. Vascular glutathione is decreased in diabetes mellitus and atherosclerosis, and the resulting deficiency of S-nitrosoglutathione may account for the increased incidence of cardiovascular complications in these conditions. 3. Tyrosine nitration—NO undergoes both oxidative and reductive reactions, resulting in a variety of oxides of nitrogen that can nitrosylate thiols and add nitrate to tyrosines (described below) or are stable oxidation products (Table 19–2). NO reacts very efficiently with superoxide to form peroxynitrite (ONOO– ), a highly reactive oxidant that leads to DNA damage, nitration of tyrosine, and oxidation of cysteine to disulfides or to various sulfur oxides (SOx ). Several cellular enzymes synthesize superoxide, and the activity of these enzymes, as well as NO synthesis, is increased in numerous inflammatory and degenerative diseases, resulting in an increase in peroxynitrite levels. Numerous proteins are susceptible to peroxynitrite-catalyzed tyrosine nitration, and this irreversible modification can be associated with either activation or inhibition of protein function. Detection of tyrosine nitration in tissue is often used as a marker of excessive NO production, although a direct causal role of tyrosine nitration in the pathogenesis of any disease has not been definitively established. Peroxynitrite-mediated protein modification is mitigated by intracellular levels of glutathione, which can protect against tissue damage by scavenging peroxynitrite. Factors that regulate the biosynthesis and decomposition of glutathione may be important modulators of the toxicity of NO. TABLE 19–2 Oxides of nitrogen.

Inactivation NO is highly labile due to its rapid reaction with metals, O2 , and reactive oxygen species. NO can react with heme and hemoproteins,


including oxyhemoglobin, which oxidizes NO to nitrate. The reaction of NO with hemoglobin may also lead to S-nitrosylation of hemoglobin, resulting in transport of NO throughout the vasculature. NO is also inactivated by reaction with O2 to form nitrogen dioxide. As noted, NO reacts with superoxide, which results in the formation of the highly reactive oxidizing species, peroxynitrite. Scavengers of superoxide anion such as superoxide dismutase may protect NO, enhancing its potency and prolonging its duration of action.

PHARMACOLOGIC MANIPULATION OF NITRIC OXIDE Inhibitors of Nitric Oxide Synthesis The primary strategy to reduce NO generation in cells is to use NOS inhibitors. The majority of these inhibitors are arginine analogs that bind to the NOS arginine-binding site. Since each of the NOS isoforms has high structural similarity, most of these inhibitors do not exhibit selectivity for individual NOS isoforms. In inflammatory disorders and sepsis (see below), inhibition of the iNOS isoform is potentially beneficial, whereas in neurodegenerative conditions, nNOS-specific inhibitors may be useful. However, administration of nonselective NOS inhibitors leads to concurrent inhibition of eNOS, which impairs its homeostatic signaling and also results in vasoconstriction and potential ischemic damage. Thus, NOS isoform-selective inhibitors are being designed that exploit subtle differences in substrate binding sites between the isoforms, as well as newer isoform-selective inhibitors that prevent NOS dimerization, the conformation required for enzymatic activity. The efficacy of NOS isoform-selective inhibitors in medical conditions is under investigation.

Nitric Oxide Donors NO donors, which release NO or related NO species, are used clinically to elicit smooth muscle relaxation. Different classes of NO donors have differing biologic properties, depending on the nature of the NO species released and the mechanism that is responsible for its release. 1. Organic nitrates—Nitroglycerin, which dilates veins and coronary arteries, is metabolized to NO by mitochondrial aldehyde reductase, an enzyme enriched in venous smooth muscle, accounting for the potent venodilating activity of this molecule. Venous dilation decreases cardiac preload, which along with coronary artery dilation accounts for the antianginal effects of nitroglycerin. Other organic nitrates, such as isosorbide dinitrate, are metabolized to an NO-releasing species through a poorly understood enzymatic pathway. Unlike NO, organic nitrates have less significant effects on aggregation of platelets, which appear to lack the enzymatic pathways necessary for rapid metabolic activation. Organic nitrates exhibit rapid tolerance during continuous administration. This nitrate tolerance may derive from the generation of reactive oxygen species that inhibit mitochondrial aldehyde reductase, endogenous NO synthesis, and other pathways (see Chapter 12). 2. Organic nitrites—Organic nitrites, such as the antianginal inhalant amyl nitrite, also require metabolic activation to elicit vasorelaxation, although the responsible enzyme has not been identified. Nitrites are arterial vasodilators and do not exhibit the rapid tolerance seen with nitrates. Amyl nitrite is abused for euphoric effects and combining it with phosphodiesterase inhibitors, such as sildenafil, can cause lethal hypotension. Amyl nitrite has been largely replaced by nitrates, such as nitroglycerin, which are more easily administered. 3. Sodium nitroprusside—Sodium nitroprusside, which dilates arterioles and venules, is used for rapid pressure reduction in arterial hypertension. In response to light as well as chemical or enzymatic mechanisms in cell membranes, sodium nitroprusside breaks down to generate five cyanide molecules and a single NO. See Chapter 11 for additional details. 4. NO gas inhalation—NO itself can be used therapeutically. Inhalation of NO results in reduced pulmonary artery pressure and improved perfusion of ventilated areas of the lung. Inhaled NO is used for pulmonary hypertension, acute hypoxemia, and cardiopulmonary resuscitation, and there is evidence of short-term improvements in pulmonary function. Inhaled NO is stored as a compressed gas mixture with nitrogen, which does not readily react with NO, and further diluted to the desired concentration upon administration. NO can react with O2 to form nitrogen dioxide, a pulmonary irritant that can cause deterioration of lung function (see Chapter 56). Additionally, NO can induce the formation of methemoglobin, a form of hemoglobin containing Fe3+ rather than Fe2+, which does not bind O2 (see also Chapter 12). Therefore, nitrogen dioxide and methemoglobin levels are monitored during inhaled NO treatment. 5. Alternate strategies—Another mechanism to potentiate the actions of NO is to inhibit the phosphodiesterase enzymes that degrade cGMP. Inhibitors of type 5 phosphodiesterase such as sildenafil result in prolongation of the duration of NO-induced cGMP elevations in a variety of tissues (see Chapter 12).


NITRIC OXIDE IN DISEASE VASCULAR EFFECTS NO has a significant effect on vascular smooth muscle tone and blood pressure. Numerous endothelium-dependent vasodilators, such as acetylcholine and bradykinin, act by increasing intracellular calcium levels in endothelial cells, leading to the synthesis of NO. NO diffuses to vascular smooth muscle leading to vasorelaxation (Figure 19–2). Mice with a knockout mutation in the eNOS gene display increased vascular tone and elevated mean arterial pressure, indicating that eNOS is a fundamental regulator of blood pressure.

FIGURE 19–2 Regulation of vasorelaxation by endothelial-derived nitric oxide (NO). Endogenous vasodilators, eg, acetylcholine and bradykinin, cause calcium (Ca2+) efflux from the endoplasmic reticulum in the luminal endothelial cells into the cytoplasm. Calcium binds to calmodulin (CaM), which activates endothelial NO synthase (eNOS), resulting in NO synthesis from l-arginine. NO diffuses into smooth muscle cells, where it activates soluble guanylyl cyclase and cyclic guanosine monophosphate (cGMP) synthesis from guanosine triphosphate (GTP). cGMP binds and activates protein kinase G (PKG), resulting in an overall reduction in calcium influx, and inhibition of calcium-dependent muscle contraction. PKG can also block other pathways that lead to muscle contraction. cGMP signaling is terminated by phosphodiesterases, which convert cGMP to GMP. Apart from being a vasodilator and regulating blood pressure, NO also has antithrombotic effects. Both endothelial cells and platelets contain eNOS, which acts via an NO-cGMP pathway to inhibit platelet activation, an initiator of thrombus formation. Thus, in diseases associated with endothelial dysfunction, the associated decrease in NO generation leads to an increased propensity for abnormal platelet function and thrombosis. NO may have an additional inhibitory effect on blood coagulation by enhancing fibrinolysis via an effect on plasminogen. NO also protects against atherogenesis. A major antiatherogenic mechanism of NO involves the inhibition of proliferation and migration of vascular smooth muscle cells. In animal models, myointimal proliferation following angioplasty can be blocked by NO donors, by NOS gene transfer, and by NO inhalation. NO reduces endothelial adhesion of monocytes and leukocytes, which are early steps in the development of atheromatous plaques. This effect is due to the inhibitory effect of NO on the expression of adhesion molecules on the endothelial surface. In addition, NO may act as an antioxidant, blocking the oxidation of low-density lipoproteins and thus preventing


or reducing the formation of foam cells in the vascular wall. Plaque formation is also affected by NO-dependent reduction in endothelial cell permeability to lipoproteins. The importance of eNOS in cardiovascular disease is supported by experiments showing increased atherosclerosis in animals treated with eNOS inhibitors. Atherosclerosis risk factors, such as smoking, hyperlipidemia, diabetes, and hypertension, are associated with decreased endothelial NO production, and thus enhance atherogenesis.

SEPTIC SHOCK Sepsis is a systemic inflammatory response caused by infection. Endotoxin components from the bacterial wall along with endogenously generated tumor necrosis factor-α and other cytokines induce synthesis of iNOS in macrophages, neutrophils, and T cells, as well as hepatocytes, smooth muscle cells, endothelial cells, and fibroblasts. This widespread generation of NO results in exaggerated hypotension, shock, and, in some cases, death. This hypotension is reduced or reversed by NOS inhibitors in humans as well as in animal models (Table 19–3). A similar reversal of hypotension is produced by compounds that prevent the action of NO, such as the sGC inhibitor methylene blue. Furthermore, knockout mice lacking a functional iNOS gene are more resistant to endotoxin than wild-type mice. However, despite the ability of NOS inhibitors to ameliorate hypotension in sepsis, there is no overall improvement in survival in patients with gram-negative sepsis treated with NOS inhibitors. The absence of benefit may reflect the inability of the NOS inhibitors used in these trials to differentiate between NOS isoforms, or may reflect concurrent inhibition of beneficial aspects of iNOS signaling. TABLE 19–3 Some inhibitors of nitric oxide synthesis or action.

INFECTION & INFLAMMATION The generation of NO has both beneficial and detrimental roles in the host immune response and in inflammation. The host response to infection or injury involves the recruitment of leukocytes and the release of inflammatory mediators, such as tumor necrosis factor and interleukin-1. This leads to induction of iNOS in leukocytes, fibroblasts, and other cell types. The NO that is produced, along with


peroxynitrite that forms from its interaction with superoxide, is an important microbicide. NO also appears to play an important protective role in the body via immune cell function. When challenged with foreign antigens, Th1 cells (see Chapter 55) respond by synthesizing NO, which has roles in Th1 cells. The importance of NO in Th1 cell function is demonstrated by the impaired protective response to injected parasites in animal models after inhibition of iNOS. NO also stimulates the synthesis of inflammatory prostaglandins by activating cyclooxygenase isoenzyme 2 (COX-2). Through its effects on COX-2, its direct vasodilatory effects, and other mechanisms, NO generated during inflammation contributes to the erythema, vascular permeability, and subsequent edema associated with acute inflammation. However, in both acute and chronic inflammatory conditions, prolonged or excessive NO production may exacerbate tissue injury. Indeed, psoriasis lesions, airway epithelium in asthma, and inflammatory bowel lesions in humans all demonstrate elevated levels of NO and iNOS, suggesting that persistent iNOS induction may contribute to disease pathogenesis. Moreover, these tissues also exhibit increased levels of nitrotyrosine, indicating excessive formation of peroxynitrite. In several animal models of arthritis, increasing NO production by dietary l-arginine supplementation exacerbates arthritis, whereas protection is seen with iNOS inhibitors. Thus, inhibition of the NO pathway may have a beneficial effect on a variety of acute and chronic inflammatory diseases.

THE CENTRAL NERVOUS SYSTEM NO has an important role in the central nervous system as a neurotransmitter (see Chapter 21). Unlike classic transmitters such as glutamate or dopamine, which are stored in synaptic vesicles and released in the synaptic cleft upon vesicle fusion, NO is not stored, but rather is synthesized on demand and immediately diffuses to neighboring cells. NO synthesis is induced at postsynaptic sites in neurons, most commonly upon activation of the NMDA subtype of glutamate receptor, which results in calcium influx and activation of nNOS. In several neuronal subtypes, eNOS is also present and activated by neurotransmitter pathways that lead to calcium influx. NO synthesized postsynaptically may function as a retrograde messenger and diffuse to the presynaptic terminal to enhance the efficiency of neurotransmitter release, thereby regulating synaptic plasticity, the process of synapse strengthening that underlies learning and memory. Because aberrant NMDA receptor activation and excessive NO synthesis is linked to excitotoxic neuronal death in several neurologic diseases, including stroke, amyotrophic lateral sclerosis, and Parkinson’s disease, therapy with NOS inhibitors may reduce neuronal damage in these conditions. However, clinical trials have not clearly supported the benefit of NOS inhibition, which may reflect nonselectivity of the inhibitors, resulting in inhibition of the beneficial effects of eNOS.

THE PERIPHERAL NERVOUS SYSTEM Nonadrenergic, noncholinergic (NANC) neurons are widely distributed in peripheral tissues, especially the gastrointestinal and reproductive tracts (see Chapter 6). Considerable evidence implicates NO as a mediator of certain NANC actions, and some NANC neurons appear to release NO. Penile erection is thought to be caused by the release of NO from NANC neurons; NO promotes relaxation of the smooth muscle in the corpora cavernosa—the initiating factor in penile erection—and inhibitors of NOS have been shown to prevent erection caused by pelvic nerve stimulation in the rat. An established approach in treating erectile dysfunction is to enhance the effect of NO signaling by inhibiting the breakdown of cGMP by the phosphodiesterase (PDE isoform 5) present in the smooth muscle of the corpora cavernosa with drugs such as sildenafil, tadalafil, and vardenafil (see Chapter 12).

RESPIRATORY DISORDERS NO is administered by inhalation to newborns with hypoxic respiratory failure associated with pulmonary hypertension. The current treatment for severely defective gas exchange in the newborn is with extracorporeal membrane oxygenation (ECMO), which does not directly affect pulmonary vascular pressures. NO inhalation dilates pulmonary vessels, resulting in decreased pulmonary vascular resistance and reduced pulmonary artery pressure. Inhaled NO also improves oxygenation by reducing mismatch of ventilation and perfusion in the lung. Inhalation of NO results in dilation of pulmonary vessels in areas of the lung with better ventilation, thereby redistributing pulmonary blood flow away from poorly ventilated areas. NO inhalation does not typically exert pronounced effects on the systemic circulation. Inhaled NO has also been shown to improve cardiopulmonary function in adult patients with pulmonary artery hypertension. An additional approach for treating pulmonary hypertension is to potentiate the actions of NO in pulmonary vascular beds. Due to the enrichment of PDE-5 in pulmonary vascular beds, PDE-5 inhibitors such as sildenafil and tadalafil induce vasodilation and marked reductions in pulmonary hypertension (see also Chapters 12 and 17).

SUMMARY Nitric Oxide


PREPARATIONS AVAILABLE

REFERENCES Chen Z, Stamler JS: Bioactivation of nitroglycerin by the mitochondrial aldehyde dehydrogenase. T rends Cardiovasc Med 2006;16:259. Griffiths MJ, Evans T W: Inhaled nitric oxide therapy in adults. N Engl J Med 2005;353:2683. Guix FX et al: T he physiology and pathophysiology of nitric oxide in the brain. Prog Neurobiol 2005;76:126. McMillan K et al: Allosteric inhibitors of inducible nitric oxide synthase dimerization discovered via combinatorial chemistry. Proc Nat Acad Sci USA 2000;97:1506. Moncada S, Higgs EA: T he discovery of nitric oxide and its role in vascular biology. Br J Pharmacol 2006;147:S193. Napoli C, Ignarro LJ: Nitric oxide-releasing drugs. Annu Rev Pharmacol T oxicol 2003;43:97. Paige JS, Jaffrey, SR: Pharmacologic manipulation of nitric oxide signaling: T argeting NOS dimerization and protein-protein interactions. Curr T opics Med Chem 2007;7:97. Wimalawansa SJ: Nitric oxide: New evidence for novel therapeutic indications. Expert Opin Pharmacother 2008;9:1935.


CHAPTER

20 Drugs Used in Asthma Joshua M. Galanter, MD, & Homer A. Boushey, MD

CASE STUDY A 10-year-old girl with a history of poorly controlled asthma presents to the emergency department with severe shortness of breath and audible inspiratory and expiratory wheezing. She is pale, refuses to lie down, and appears extremely frightened. Her pulse is 120 bpm and respirations 32/min. Her mother states that the girl has just recovered from a mild case of flu and had seemed comfortable until this afternoon. The girl uses an inhaler (albuterol) but “only when really needed” because her parents are afraid that she will become too dependent on medication. She administered two puffs from her inhaler just before coming to the hospital, but “the inhaler doesn’t seem to have helped.” What emergency measures are indicated? How should her long-term management be altered?

A consistent increase in the prevalence of asthma over the past 60 years has made it an extraordinarily common disease. The reasons for this increase—shared across all modern, “westernized” societies—are poorly understood, but in the United States alone, 18.9 million adults and 7.1 million children currently have asthma. The condition accounts for 15 million outpatient visits, 1.8 million emergency department visits, and 440,000 hospitalizations each year. Despite substantial improvements in the treatment for the disease, asthma still accounts for 3400 deaths per year in the USA. The clinical features of asthma are recurrent bouts of shortness of breath, chest tightness, and wheezing, often associated with coughing. Its hallmark physiologic features are widespread, reversible narrowing of the bronchial airways and a marked increase in bronchial responsiveness to inhaled stimuli; and its pathologic features are lymphocytic, eosinophilic inflammation of the bronchial mucosa. These changes are accompanied by “remodeling” of the bronchial wall, with thickening of the lamina reticularis beneath the epithelium and hyperplasia of the bronchial vasculature, smooth muscle, secretory glands, and goblet cells. In mild asthma, symptoms occur only intermittently, as on exposure to allergens or air pollutants, on exercise, or after viral upper respiratory infection. More severe forms of asthma are associated with more frequent and severe symptoms, especially at night. Chronic airway constriction causes persistent respiratory impairment, punctuated by frequent acute asthmatic attacks, or “asthma exacerbations.” These attacks are most often associated with viral respiratory infections and are characterized by severe airflow obstruction from intense contraction of airway smooth muscle, inspissation of mucus plugs in the airway lumen, and thickening of the bronchial mucosa from edema and inflammatory cell infiltration. The spectrum of asthma’s severity is wide, and patients are classified as having “mild intermittent,” “mild persistent,” “moderate persistent,” and “severe persistent,” either based on the frequency and severity of symptoms and the severity of airflow obstruction on pulmonary function testing or by the minimal medical therapy required to keep their asthma well-controlled, and as “exacerbation-prone” or “exacerbation-resistant” based on the frequency of asthma exacerbations. Until recently, the entire range of asthma severity was regarded as eminently treatable, because treatments for quick relief of symptoms of acute bronchoconstriction (“short-term relievers”) and treatments for reduction in symptoms and prevention of attacks (“long-term controllers”) have been shown effective in many large, well-designed clinical trials, case-control studies, and evidence-based reviews. The persistence of high medical costs for asthma, driven largely by the costs of emergency department and hospital treatment of asthma exacerbations, was thus believed to reflect underutilization of the treatments available. Reconsideration of this view was driven by recognition that the term “asthma” is applied to a variety of different disorders sharing common clinical features but fundamentally different pathophysiologic mechanisms. Attention has thus turned to the possibility that there are different asthma forms or phenotypes, some of which are less responsive to the current mainstays of asthma controller therapy. The current view of asthma treatment may be summarized as follows: that the treatments commonly used at present are indeed effective for the most common form of the disease, as it presents in children and young adults with allergic asthma, but that there are other phenotypes of asthma for which these therapies are less effective, and that represent an unmet medical need. Accordingly, this chapter first reviews the pathophysiology of the most common form of asthma and the basic pharmacology of the agents used in its treatment. It will then turn to discussion of different forms


or phenotypes of asthma and the efforts to develop effective therapies for them.

PATHOGENESIS OF ASTHMA Classic allergic asthma is regarded as mediated by immune globulin (IgE), produced in response to exposure to foreign proteins, like those from house dust mite, cockroach, animal danders, molds, and pollens. These qualify as allergens on the basis of their induction of IgE antibody production in people exposed to them. The tendency to produce IgE is at least in part genetically determined, and asthma clusters with other allergic diseases (allergic rhinitis, eczema) in family groups. Once produced, IgE binds to high-affinity receptors (FcεR-1) on mast cells in the airway mucosa (Figure 20–1), so that re-exposure to the allergen triggers the release of mediators stored in the mast cells’ granules and the synthesis and release of other mediators. The histamine, tryptase, leukotrienes C4 and D4 , and prostaglandin D2 released cause the smooth muscle contraction and vascular leakage responsible for the acute bronchoconstriction of the “early asthmatic response.” This response is often followed in 3–6 hours by a second, more sustained phase of bronchoconstriction, the “late asthmatic response,” associated with an influx of inflammatory cells into the bronchial mucosa and with an increase in bronchial reactivity. This late response is thought to be due to cytokines characteristically produced by T H2 lymphocytes, especially interleukins 5, 9, and 13. These cytokines are thought to attract and activate eosinophils, stimulate IgE production by B lymphocytes, and stimulate mucus production by bronchial epithelial cells. It is not clear whether lymphocytes or mast cells in the airway mucosa are the primary source of the mediators responsible for the late inflammatory response, but the benefits of corticosteroid therapy are attributed to their inhibition of the production of pro-inflammatory cytokines in the airways.


FIGURE 20–1 Conceptual model for the immunopathogenesis of asthma. Exposure to allergen causes synthesis of IgE, which binds to mast cells in the airway mucosa. On re-exposure to allergen, antigen-antibody interaction on mast cell surfaces triggers release of mediators of anaphylaxis: histamine, tryptase, prostaglandin D2 (PGD2 ), leukotriene (LT) C4 , and platelet-activating factor (PAF). These agents provoke contraction of airway smooth muscle, causing the immediate fall in forced expiratory volume in 1 sec (FEV1 ). Reexposure to allergen also causes the synthesis and release of a variety of cytokines: interleukins (IL) 4 and 5, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor (TNF), and tissue growth factor (TGF) from T cells and mast cells. These cytokines in turn attract and activate eosinophils and neutrophils, whose products include eosinophil cationic protein (ECP), major basic protein (MBP), proteases, and platelet-activating factor. These mediators cause the edema, mucus hypersecretion, smooth muscle contraction, and increase in bronchial reactivity associated with the late asthmatic response, indicated by a second fall in FEV1 3–6 hours after the exposure.


The allergen challenge model does not reproduce all the features of asthma. Most asthma attacks are not triggered by inhalation of allergens, but instead by viral respiratory infections. Some adults with asthma have no evidence of allergic sensitivity to allergens, and bronchospasm can be provoked by nonallergenic stimuli such as distilled water aerosol, exercise, cold air, cigarette smoke, and sulfur dioxide. This tendency to develop bronchospasm on encountering nonallergenic stimuli—assessed by measuring the fall in maximal expiratory flow provoked by inhaling serially increasing concentrations of the aerosolized cholinergic agonist methacholine—is described as “bronchial hyper-reactivity.” It is considered fundamental to asthma’s pathogenesis because it is nearly ubiquitous in patients with asthma, and its degree roughly correlates with the clinical severity of the disease. The mechanisms underlying bronchial hyper-reactivity are incompletely understood but appear to be related to inflammation of the airway mucosa. The anti-inflammatory activity of inhaled corticosteroid (ICS) treatment is credited with preventing the increase in reactivity associated with the late asthmatic response (Figure 20–1). Whatever the mechanisms responsible for bronchial hyper-reactivity, bronchoconstriction itself results not simply from the direct effect of the released mediators but also from their activation of neural pathways. This is suggested by the effectiveness of muscarinic receptor antagonists, which have no direct effect on smooth muscle contractility, in inhibiting the bronchoconstriction caused by inhalation of allergens and airway irritants. The hypothesis suggested by this conceptual model—that asthmatic bronchospasm results from a combination of release of mediators and an exaggeration of responsiveness to their effects—predicts that drugs with different modes of action may effectively treat asthma. Asthmatic bronchospasm might be reversed or prevented, for example, by drugs that reduce the amount of IgE bound to mast cells (antiIgE antibody), prevent mast cell degranulation (cromolyn or nedocromil, sympathomimetic agents, calcium channel blockers), block the action of the products released (antihistamines and leukotriene receptor antagonists), inhibit the effect of acetylcholine released from vagal motor nerves (muscarinic antagonists), or directly relax airway smooth muscle (sympathomimetic agents, theophylline). The second approach to the treatment of asthma is aimed at reducing the level of bronchial responsiveness. Because increased responsiveness appears to be linked to airway inflammation and because airway inflammation is a feature of late asthmatic responses, this strategy is implemented both by reducing exposure to the allergens that provoke inflammation and by prolonged therapy with antiinflammatory agents, especially inhaled corticosteroids (ICS).

BASIC PHARMACOLOGY OF AGENTS USED IN THE TREATMENT OF ASTHMA The drugs most used for management of asthma are adrenoceptor agonists, or sympathomimetic agents (used as “relievers” or bronchodilators) and inhaled corticosteroids (used as “controllers” or anti-inflammatory agents). Their basic pharmacology is presented elsewhere (see Chapters 9 and 39). In this chapter, we review their pharmacology relevant to asthma.

SYMPATHOMIMETIC AGENTS Adrenoceptor agonists are mainstays in the treatment of asthma. Their binding to β receptors—abundant on airway smooth muscle cells —stimulates adenylyl cyclase and increases the formation of intracellular cAMP (Figure 20–2), thereby relaxing airway smooth muscle and inhibiting release of bronchoconstricting mediators from mast cells. They may also inhibit microvascular leakage and increase mucociliary transport. Adverse effects, especially of adrenoceptor agonists that activate β 1 as well as β2 receptors, include tachycardia, skeletal muscle tremor, and decreases in serum potassium levels.


FIGURE 20–2 Bronchodilation is promoted by cAMP. Intracellular levels of cAMP can be increased by β-adrenoceptor agonists, which increase the rate of its synthesis by adenylyl cyclase (AC); or by phosphodiesterase (PDE) inhibitors such as theophylline, which slow the rate of its degradation. Bronchoconstriction can be inhibited by muscarinic antagonists and possibly by adenosine antagonists. Sympathomimetic agents that have been widely used in the treatment of asthma include epinephrine, ephedrine, isoproterenol, and albuterol and other β2 -selective agents (Figure 20–3). Because epinephrine and isoproterenol increase the rate and force of cardiac contraction (mediated mainly by β1 receptors), they are reserved for special situations (see below).


FIGURE 20–3 Structures of isoproterenol and several β2 -selective analogs. In general, adrenoceptor agonists are best delivered by inhalation. This results in the greatest local effect on airway smooth muscle with the least systemic toxicity. Aerosol deposition depends on the particle size, the pattern of breathing, and the geometry of the airways. Even with particles in the optimal size range of 2–5 mm, 80–90% of the total dose of aerosol is deposited in the mouth or pharynx. Particles under 1–2 μm remain suspended and may be exhaled. Bronchial deposition of an aerosol is increased by slow inhalation of a nearly full breath and by 5 or more seconds of breath-holding at the end of inspiration. Epinephrine is an effective, rapidly acting bronchodilator when injected subcutaneously (0.4 mL of 1:1000 solution) or inhaled as a microaerosol from a pressurized canister (320 mcg per puff). Maximal bronchodilation is achieved 15 minutes after inhalation and lasts 60–90 minutes. Because epinephrine stimulates α and β1 as well as β2 receptors, tachycardia, arrhythmias, and worsening of angina pectoris are troublesome adverse effects. The cardiovascular effects of epinephrine are of value for treating the acute vasodilation and shock as well as the bronchospasm of anaphylaxis, but other, more β2 -selective agents have displaced its use in asthma. Ephedrine was used in China for more than 2000 years before its introduction into Western medicine in 1924. Compared with epinephrine, ephedrine has a longer duration, oral activity, more pronounced central effects, and much lower potency. Because of the development of more efficacious and β2 -selective agonists, ephedrine is now used infrequently in treating asthma. Isoproterenol is a potent nonselective β1 and β2 bronchodilator. When inhaled as a microaerosol from a pressurized canister, 80– 120 mcg isoproterenol causes maximal bronchodilation within 5 minutes and has a 60- to 90-minute duration of action. An increase in asthma mortality in the United Kingdom in the mid-1960s was attributed to cardiac arrhythmias resulting from the use of high doses of inhaled isoproterenol. It is now rarely used for asthma.


Beta2-Selective Drugs The β2 -selective adrenoceptor agonist drugs, particularly albuterol, are now the most widely used sympathomimetics for the treatment of the bronchoconstriction of asthma (Figure 20–3). These agents differ structurally from epinephrine in having a larger substitution on the amino group and in the position of the hydroxyl groups on the aromatic ring. They are effective after inhaled or oral administration and have a longer duration of action than epinephrine or isoproterenol. Albuterol, terbutaline, metaproterenol, and pirbuterol are available as metered-dose inhalers. Given by inhalation, these agents cause bronchodilation equivalent to that produced by isoproterenol. Bronchodilation is maximal within 15 minutes and persists for 3–4 hours. All can be diluted in saline for administration from a hand-held nebulizer. Because the particles generated by a nebulizer are much larger than those from a metered-dose inhaler, much higher doses must be given (2.5–5.0 mg vs 100–400 mcg) but are no more effective. Nebulized therapy should thus be reserved for patients unable to coordinate inhalation from a metered-dose inhaler. Most preparations of β2 -selective drugs are a mixture of R and S isomers. Only the R isomer activates the β-agonist receptor. Reasoning that the S isomer may promote inflammation, a purified preparation of the R isomer of albuterol has been developed (levalbuterol). Whether this actually presents significant advantages in clinical use is still unproven. Albuterol and terbutaline are also available in oral form. One tablet two or three times daily is the usual regimen; the principal adverse effects are skeletal muscle tremor, nervousness, and occasional weakness. This route of administration presents no advantage over inhaled treatment and is rarely prescribed. Of these agents, only terbutaline is available for subcutaneous injection (0.25 mg). The indications for this route are similar to those for subcutaneous epinephrine—severe asthma requiring emergency treatment when aerosolized therapy is not available or has been ineffective—but it should be remembered that terbutaline’s longer duration of action means that cumulative effects may be seen after repeated injections. Large doses of parenteral terbutaline are sometimes used to inhibit the uterine contractions associated with premature labor. A newer generation of long-acting β 2 -selective agonists includes salmeterol (a partial agonist) and formoterol (a full agonist). These long-acting β agonists (LABA) are potent selective β2 agonists that achieve their long duration of action (12 hours or more) as a result of high lipid solubility. This permits them to dissolve in the smooth muscle cell membrane in high concentrations or, possibly, attach to “mooring” molecules in the vicinity of the adrenoceptor. These drugs appear to interact with inhaled corticosteroids to improve asthma control. Because they have no anti-inflammatory action, they should not be used as monotherapy for asthma. Ultra-long-acting β agonists, indacaterol, olodaterol, and vilanterol, need to be taken only once a day but are currently FDA-approved only for the treatment of chronic obstructive pulmonary disease (COPD). Other long-acting β agonists approved in Europe, but not yet in the United States include bambuterol.

Toxicities Concerns over the potential toxicities of acute treatment of asthma with inhaled sympathomimetic agents—worsened hypoxemia and cardiac arrhythmia—have been largely put to rest. It is true that the vasodilating action of β2 -agonist treatment may increase perfusion of poorly ventilated lung units, transiently decreasing arterial oxygen tension (PaO2 ), but this effect is small, is easily overcome by the routine administration of supplemental oxygen in the treatment of severe attacks of asthma, and is soon made irrelevant by the increase in oxygen tension that follows β-agonist-induced bronchodilation. The other concern, precipitation of cardiac arrhythmias, appears unsubstantiated. In patients presenting for emergency treatment of severe asthma, irregularities in cardiac rhythm improve with the improvements in gas exchange effected by bronchodilator treatment and oxygen administration. Not all of the concerns over the potential toxicities of chronic treatment with an inhaled β agonist—made easy by the introduction of long-acting β agonists—have been as easily resolved. One that has been resolved is the induction of tachyphylaxis to their bronchodilator action. A reduction in the bronchodilator response to low-dose β-agonist treatment can indeed be shown after several days of regular βagonist use, but maximal bronchodilation is still achieved well within the range of doses usually given. Tachyphylaxis is more clearly reflected in a loss of the protection afforded by acute treatment with a β agonist against a later challenge by exercise or inhalation of allergen or an airway irritant. It remains to be demonstrated in a clinical trial, however, whether this loss of bronchoprotective efficacy is associated with adverse outcomes. The demonstration of genetic variations in the β receptor raised the possibility that the risks of adverse effects might not be uniformly distributed among asthmatic patients. Attention has focused on the receptor’s B16 locus. Retrospective analyses of studies of regular βagonist treatment suggested that asthma control deteriorated among patients homozygous for arginine at this locus, a genotype found in 16% of the Caucasian population but more commonly in African Americans. It was thus tempting to speculate that a genetic variant may underlie the report of an increase in asthma mortality from regular use of a long-acting β agonist in studies involving very large numbers of patients (see below), but several studies of LABA treatment have since shown the differences in multiple measures of asthma control to be nil or very small in asthmatics with different Arg/Gly variations at the B16 locus. One large study of COPD patients has even suggested that regular use of salmeterol reduced the risk of exacerbations in patients homozygous for arginine at the B16 locus. The importance of genetic variants in the gene for the B16 locus in the β receptor is thus uncertain. It is nonetheless certain that


pharmacogenetic studies of asthma treatment will continue to be an active focus of research, as an approach to the development of “personalized therapy.”

METHYLXANTHINE DRUGS The three important methylxanthines are theophylline, theobromine, and caffeine. Their major source is beverages (tea, cocoa, and coffee, respectively). The use of theophylline, once a mainstay of asthma treatment, has waned with demonstration of the greater efficacy of inhaled adrenoceptor agonists for acute asthma and of inhaled anti-inflammatory agents for chronic asthma. Accelerating this decline in theophylline’s use are its toxicities (nausea, vomiting, tremulousness, arrhythmias) and the requirement for monitoring serum levels because of the narrowness of its therapeutic index. This monitoring is made all the more necessary by individual and drugassociated differences in theopylline metabolism.

Chemistry As shown below (Figure 20–4), theophylline is 1,3-dimethylxanthine; theobromine is 3,7-dimethylxanthine; and caffeine is 1,3,7trimethylxanthine. A theophylline preparation commonly used for therapeutic purposes is aminophylline, a theophylline-ethylenediamine complex. The pharmacokinetics of theophylline are discussed below (see Clinical Uses of Methylxanthines). Its metabolic products, partially demethylated xanthines (not uric acid), are excreted in the urine.

FIGURE 20–4 Structures of theophylline and other methylxanthines.

Mechanism of Action Several mechanisms have been proposed for the actions of methylxanthines, but none has been firmly established. At high concentrations, they can be shown in vitro to inhibit several members of the phosphodiesterase (PDE) enzyme family thereby increasing concentrations of intracellular cAMP and, in some tissues, cGMP (Figure 20–2). Cyclic AMP regulates many cellular functions including, but not limited to, stimulation of cardiac function, relaxation of smooth muscle, and reduction in the immune and inflammatory activity of specific cells. Of the various isoforms of PDE identified, inhibition of PDE3 appears to be the most involved in relaxing airway smooth muscle and inhibition of PDE4 in inhibiting release of cytokines and chemokines, which in turn results in a decrease in immune cell migration and


activation. This anti-inflammatory effect is achieved at doses lower than those necessary for bronchodilation. In an effort to reduce toxicity while maintaining therapeutic efficacy, selective inhibitors of PDE4 have been developed. Many were abandoned after clinical trials showed that their toxicities of nausea, headache, and diarrhea restricted dosing to subtherapeutic levels, but one, roflumilast, has been approved by the FDA as a treatment for COPD, though not for asthma. Another proposed mechanism is inhibition of cell surface receptors for adenosine. These receptors modulate adenylyl cyclase activity, and adenosine has been shown to provoke contraction of isolated airway smooth muscle and histamine release from airway mast cells. It has been shown, however, that xanthine derivatives devoid of adenosine antagonism (eg, enprofylline) can inhibit bronchoconstriction in asthmatic subjects. A third mechanism of action may underlie theophylline’s efficacy: enhancement of histone deacetylation. Acetylation of core histones is necessary for activation of inflammatory gene transcription. Corticosteroids act, at least in part, by recruiting histone deacetylases to the site of inflammatory gene transcription, an action enhanced by low-dose theophylline. This interaction should predict that low-dose theophylline treatment would enhance the effectiveness of corticosteroid treatment, and some clinical trials indeed support the idea that theophylline treatment is effective as add-on therapy in patients with asthma or COPD uncontrolled by ICS plus LABA therapy.

Pharmacodynamics The methylxanthines have effects on the central nervous system, kidney, and cardiac and skeletal muscle as well as smooth muscle. Of the three agents, theophylline is most selective in its smooth muscle effects, whereas caffeine has the most marked central nervous system effects. A. Central Nervous System Effects All methylxanthines—but especially caffeine—cause mild cortical arousal with increased alertness and deferral of fatigue. The caffeine contained in beverages—eg, 100 mg in a cup of coffee—is sufficient to cause nervousness and insomnia in sensitive individuals and slight bronchodilation in patients with asthma. The larger doses necessary for more effective bronchodilation cause nervousness and tremor. Very high doses, from accidental or suicidal overdose, cause medullary stimulation, convulsions, and even death. B. Cardiovascular Effects Methylxanthines have positive chronotropic and inotropic effects on the heart. At low concentrations, these effects result from inhibition of presynaptic adenosine receptors in sympathetic nerves, increasing catecholamine release at nerve endings. The higher concentrations (> 10 μmol/L, 2 mg/L) associated with inhibition of phosphodiesterase and increases in cAMP may result in increased influx of calcium. At much higher concentrations (> 100 μmol/L), sequestration of calcium by the sarcoplasmic reticulum is impaired. The clinical expression of these effects on cardiovascular function varies among individuals. Ordinary consumption of methylxanthine-containing beverages usually produces slight tachycardia, an increase in cardiac output, and an increase in peripheral resistance, raising blood pressure slightly. In sensitive individuals, consumption of a few cups of coffee may result in arrhythmias. High doses of these agents relax vascular smooth muscle except in cerebral blood vessels, where they cause contraction. Methylxanthines decrease blood viscosity and may improve blood flow under certain conditions. The mechanism of this action is not well defined, but the effect is exploited in the treatment of intermittent claudication with pentoxifylline, a dimethylxanthine agent. C. Effects on Gastrointestinal Tract The methylxanthines stimulate secretion of both gastric acid and digestive enzymes. However, even decaffeinated coffee has a potent stimulant effect on secretion, which means that the primary secretagogue in coffee is not caffeine. D. Effects on Kidney The methylxanthines—especially theophylline—are weak diuretics. This effect may involve both increased glomerular filtration and reduced tubular sodium reabsorption. The diuresis is not of sufficient magnitude to be therapeutically useful. E. Effects on Smooth Muscle The bronchodilation produced by the methylxanthines is the major therapeutic action in asthma. Tolerance does not develop, but adverse effects, especially in the central nervous system, limit the dose (see below). In addition to their effect on airway smooth muscle, these agents—in sufficient concentration—inhibit antigen-induced release of histamine from lung tissue; their effect on mucociliary transport is unknown. F. Effects on Skeletal Muscle The respiratory actions of methylxanthines are not confined to the airways, for they also improve contractility of skeletal muscle and reverse fatigue of the diaphragm in patients with COPD. This effect—rather than an effect on the respiratory center—may account for


theophylline’s ability to improve the ventilatory response to hypoxia and to diminish dyspnea even in patients with irreversible airflow obstruction.

Clinical Uses Of the xanthines, theophylline is the most effective bronchodilator. It relieves airflow obstruction in acute asthma and reduces the severity of symptoms in patients with chronic asthma. Theophylline base is only slightly soluble in water, so it is administered as several salts containing varying amounts of theophylline base. Most preparations are well absorbed from the gastrointestinal tract; absorption of rectal suppositories is unreliable. Numerous sustained-release preparations are available and can produce therapeutic blood levels for 12 hours or more. These preparations offer the advantages of less frequent drug administration, less fluctuation of theophylline blood levels, and more effective treatment of nocturnal bronchospasm. Theophylline should be used only where methods to measure blood levels are available. Improvement in pulmonary function is correlated with plasma concentrations in the range of 5–20 mg/L. Anorexia, nausea, vomiting, abdominal discomfort, headache, and anxiety may occur at concentrations of 15 mg/L and become common at concentrations more than 20 mg/L. Higher levels (> 40 mg/L) may cause seizures or arrhythmias; these may not be preceded by gastrointestinal or neurologic warning symptoms. The plasma clearance of theophylline varies widely. It is metabolized by the liver, so usual doses may lead to toxic concentrations in patients with liver disease. Conversely, clearance may be increased through the induction of hepatic enzymes by cigarette smoking or by changes in diet. In normal adults, the mean plasma clearance is 0.69 mL/kg/min. Children clear theophylline faster than adults (1–1.5 mL/kg/min). Neonates and young infants have the slowest clearance (see Chapter 60). Even when maintenance doses are altered to correct for the above factors, plasma concentrations vary widely. Theophylline improves long-term control of asthma when taken as the sole maintenance treatment or when added to inhaled corticosteroids. It is inexpensive, and it can be taken orally. Its use, however, also requires occasional measurement of plasma levels; it often causes unpleasant minor side effects (especially insomnia); and accidental or intentional overdose can result in severe toxicity or death. For oral therapy with the prompt-release formulation, the usual dose is 3–4 mg/kg of theophylline every 6 hours. Changes in dosage result in a new steady-state concentration of theophylline in 1–2 days, so the dosage may be increased at intervals of 2–3 days until therapeutic plasma concentrations are achieved (10–20 mg/L) or until adverse effects develop. The development of more effective bronchodilators (β2 -selective adrenergic agonists) and more effective anti-inflammatory agents (ICS) with fewer adverse effects has resulted in the decline in the clinical use of theophylline. Typically, theophylline is rarely used as monotherapy and, when prescribed, is most commonly used as add-on therapy when treatment with other agents, principally ICS, is inadequate.

ANTIMUSCARINIC AGENTS Observation of the use of leaves from Datura stramonium for asthma treatment in India led to the discovery of atropine, a potent competitive inhibitor of acetylcholine at postganglionic muscarinic receptors, as a bronchodilator. Interest in the potential value of antimuscarinic agents increased with demonstration of the importance of the vagus nerves in bronchospastic responses of laboratory animals and with the development of ipratropium, a potent atropine analog that is poorly absorbed after aerosol administration and is therefore relatively free of systemic atropine-like effects.

Mechanism of Action Muscarinic antagonists competitively inhibit the action of acetylcholine at muscarinic receptors (see Chapter 8). In the airways, acetylcholine is released from efferent endings of the vagus nerves, and muscarinic antagonists block the contraction of airway smooth muscle and the increase in secretion of mucus that occurs in response to vagal activity (Figure 20–5). Very high concentrations—well above those achieved even with maximal therapy—are required to inhibit the response of airway smooth muscle to nonmuscarinic stimulation. This selectivity of muscarinic antagonists accounts for their usefulness as investigative tools to examine the role of parasympathetic pathways in bronchomotor responses but limits their usefulness in preventing bronchospasm. In the doses given, antimuscarinic agents inhibit only that portion of the response mediated by muscarinic receptors, which varies by stimulus, and which further appears to vary among individual responses to the same stimulus.


FIGURE 20–5 Mechanisms of response to inhaled irritants. The airway is represented microscopically by a cross-section of the wall with branching vagal sensory endings lying adjacent to the lumen. Afferent pathways in the vagus nerves travel to the central nervous system; efferent pathways from the central nervous system travel to efferent ganglia. Postganglionic fibers release acetylcholine (ACh), which binds to muscarinic receptors on airway smooth muscle. Inhaled materials may provoke bronchoconstriction by several possible mechanisms. First, they may trigger the release of chemical mediators from mast cells. Second, they may stimulate afferent receptors to initiate reflex bronchoconstriction or to release tachykinins (eg, substance P) that directly stimulate smooth muscle contraction.

Clinical Uses Antimuscarinic agents are effective bronchodilators. Even when administered by aerosol, the bronchodilation achievable with atropine, the prototypic muscarinic antagonist, is limited by absorption into the circulation and across the blood-brain barrier. Greater bronchodilation, with less toxicity from systemic absorption, is achieved by treatment with a selective quaternary ammonium derivative of atropine, ipratropium bromide. Ipratropium can be delivered in high doses by this route because it is poorly absorbed into the circulation


and does not readily enter the central nervous system. Studies with this agent have shown that the degree of involvement of parasympathetic pathways in bronchomotor responses varies among subjects. In some, bronchoconstriction is inhibited effectively; in others, only modestly. The failure of higher doses of the muscarinic antagonist to further inhibit the response in these individuals indicates that mechanisms other than parasympathetic reflex pathways must be involved. Even in the subjects least protected by this antimuscarinic agent, however, the bronchodilation and partial inhibition of provoked bronchoconstriction are of clinical value, and antimuscarinic agents are especially useful for patients intolerant of inhaled β-agonist agents. Although antimuscarinic drugs appear to be slightly less effective in reversing asthmatic bronchospasm, the addition of ipratropium enhances the bronchodilation produced by nebulized albuterol in acute severe asthma. Ipratropium appears to be as effective as albuterol in patients with COPD who have at least partially reversible obstruction. Longeracting antimuscarinic agents, tiotropium and aclidinium, are approved for maintenance therapy of COPD. These drugs bind to M1 , M2 , and M3 receptors with equal affinity, but dissociate most rapidly from M 2 receptors, expressed on the efferent nerve ending. This means that they do not inhibit the M2 -receptor-mediated inhibition of acetylcholine release and thus benefit from a degree of receptor selectivity. They are taken by inhalation. A single dose of 18 mcg of tiotropium has a 24-hour duration of action, whereas inhalation of 400 mcg of aclidinium has a 12-hour duration of action and is thus taken twice daily. Daily inhalation of tiotropium has been shown not only to improve functional capacity of patients with COPD, but also to reduce the frequency of exacerbations of their condition. Neither drug has been approved as maintenance treatment for asthma, but the addition of tiotropium has recently been shown to be no less effective than addition of a long-acting β agonist in asthmatic patients insufficiently controlled by ICS therapy alone.

CORTICOSTEROIDS Mechanism of Action Corticosteroids (specifically, glucocorticoids) have long been used in the treatment of asthma and are presumed to act by their broad antiinflammatory efficacy, mediated in part by inhibition of production of inflammatory cytokines (see Chapter 39). They do not relax airway smooth muscle directly but reduce bronchial hyper-reactivity and reduce the frequency of asthma exacerbations if taken regularly. Their effect on airway obstruction is due in part to their contraction of engorged vessels in the bronchial mucosa and their potentiation of the effects of β-receptor agonists, but their most important action is inhibition of the infiltration of asthmatic airways by lymphocytes, eosinophils, and mast cells. The remarkable benefits of glucocorticoid treatment for patients with severe asthma have been noted since the 1950s. So too, unfortunately, have been the numerous and severe toxicities of systemic glucocorticoid treatment, especially when given repeatedly, as is necessary for a chronic disease like asthma. The development of beclomethasone in the 1970s as a topically active glucocorticoid preparation that can be taken by inhalation was thus a landmark development. It enabled delivery of high doses of a glucocorticoid to the target tissue—the bronchial mucosa—with little absorption into the systemic circulation. The development of ICS has transformed the treatment of all but mild, intermittent asthma, which can be controlled by “as-needed” use of albuterol alone.

Clinical Uses Clinical studies of corticosteroids consistently show them to be effective in improving all indices of asthma control: severity of symptoms, tests of airway caliber and bronchial reactivity, frequency of exacerbations, and quality of life. Because of severe adverse effects when given chronically, oral and parenteral corticosteroids are reserved for patients who require urgent treatment, ie, those who have not improved adequately with bronchodilators or who experience worsening symptoms despite maintenance therapy. Regular or “controller” therapy is maintained with ICS in all but the most severely affected individuals. Urgent treatment is often begun with an oral dose of 30–60 mg prednisone per day or an intravenous dose of 1 mg/kg methylprednisolone every 6–12 hours; the dose is decreased after airway obstruction has improved. In most patients, systemic corticosteroid therapy can be discontinued in 5–10 days, but in other patients symptoms may worsen as the dose is decreased to lower levels. Inhalational treatment is the most effective way to avoid the systemic adverse effects of corticosteroid therapy. The introduction of ICS such as beclomethasone, budesonide, ciclesonide, flunisolide, fluticasone, mometasone, and triamcinolone has made it possible to deliver corticosteroids to the airways with minimal systemic absorption. An average daily dose of 800 mcg of inhaled beclomethasone is equivalent to about 10–15 mg/d of oral prednisone for the control of asthma, with far fewer systemic effects. Indeed, one of the cautions in switching patients from oral to ICS therapy is to taper oral therapy slowly to avoid precipitation of adrenal insufficiency. In patients requiring continued prednisone treatment despite standard doses of an inhaled corticosteroid, higher inhaled doses are often effective and enable tapering and discontinuing prednisone treatment. Although these high doses of inhaled steroids may cause adrenal suppression, the risks of systemic toxicity from their chronic use are negligible compared with those of the oral corticosteroid therapy they replace. A special problem caused by inhaled topical corticosteroids is the occurrence of oropharyngeal candidiasis. This is easily treated with topical cotrimazole, and the risk of this complication can be reduced by having patients gargle water and expectorate after each inhaled


treatment. Ciclesonide, the most recently approved ICS, is a prodrug activated by bronchial esterases, and though no more effective in the treatment of asthma, has been associated with less frequent candidiasis. Hoarseness can also result from a direct local effect of ICS on the vocal cords. Although a majority of the inhaled dose is deposited in the oropharynx and swallowed, inhaled corticosteroids are subject to first-pass metabolism in the liver and thus are remarkably free of other short-term complications in adults. Nonetheless, chronic use may increase the risks of osteoporosis and cataracts. In children, ICS therapy has been shown to slow the rate of growth by about 1 cm over the first year of treatment, but not the rate of growth thereafter, so that the effect on adult height is minimal. Because of the efficacy and safety of inhaled corticosteroids, national and international guidelines for asthma management recommend their prescription for patients who require more than occasional inhalations of a β agonist for relief of symptoms. This therapy is continued for 10–12 weeks and then withdrawn to determine whether more prolonged therapy is needed. Inhaled corticosteroids are not curative. In most patients, the manifestations of asthma return within a few weeks after stopping therapy even if they have been taken in high doses for 2 or more years. A prospective, placebo-controlled study of the early, sustained use of inhaled corticosteroids in young children with asthma showed significantly greater improvement in asthma symptoms, pulmonary function, and frequency of asthma exacerbations over the 2 years of treatment, but no difference in overall asthma control 3 months after the end of the trial. Inhaled corticosteroids are thus properly labeled as “controllers.” They are effective only so long as they are taken. Another approach to reducing the risk of long-term, twice-daily use of ICS is to administer them only intermittently, when symptoms of asthma flare. Taking a single inhalation of an ICS with each inhalation of a short-acting β-agonist reliever (eg, an inhalation of beclomethasone for each inhalation of albuterol) or taking a 5- to 10-day course of twice-daily high-dose budesonide or beclomethasone when asthma symptoms worsen has been found to be as effective as regular daily therapy in adults and children with mild to moderate asthma, although these approaches to treatment are neither endorsed by guidelines for asthma management nor approved by the FDA.

CROMOLYN & NEDOCROMIL Cromolyn sodium (disodium cromoglycate) and nedocromil sodium were once widely used for asthma management, especially in children, but have now been supplanted so completely by other therapies that they are mostly of historic interest. Both have low solubility, are poorly absorbed from the gastrointestinal tract, and must be inhaled as a microfine powder or microfine suspension. These drugs have no effect on airway smooth muscle tone and are ineffective in reversing asthmatic bronchospasm but effectively inhibit both antigen- and exercise-induced asthma.

Mechanism of Action Cromolyn and nedocromil are thought to alter the function of delayed chloride channels in cell membranes, inhibiting cell activation. This action on airway nerves is thought to mediate inhibition of cough; on mast cells and eosinophils, the drugs inhibit the early and the late response to antigen challenge.


Clinical Uses In short-term clinical trials, pretreatment with cromolyn or nedocromil blocks the bronchoconstriction caused by allergen inhalation, exercise, sulfur dioxide, and a variety of causes of occupational asthma. This acute protective effect of a single treatment makes cromolyn useful for administration shortly before exercise or before unavoidable exposure to an allergen. When taken regularly (2-4 puffs 2-4 times daily) both agents modestly but significantly reduce symptomatic severity and the need for bronchodilator medications, particularly in young patients with allergic asthma. These drugs are not as potent or as predictably effective as ICS, and the only way of determining whether a patient will respond is by a therapeutic trial of 4 weeks’ duration. Cromolyn and nedocromil solutions are also useful in reducing symptoms of allergic rhinoconjunctivitis. Applying the solution by nasal spray or eye drops several times a day is effective in about 75% of patients, even during the peak pollen season. Because the drugs are so poorly absorbed, adverse effects of cromolyn and nedocromil are minor and are localized to the sites of deposition. These include throat irritation, cough, and mouth dryness, and, rarely, chest tightness and wheezing. Inhalation of a β 2 adrenoceptor agonist before cromolyn or nedocromil treatment can prevent some of these symptoms. Serious adverse effects are rare. Reversible dermatitis, myositis, or gastroenteritis occurs in less than 2% of patients, and a very few cases of pulmonary infiltration with eosinophilia and anaphylaxis have been reported. This lack of toxicity accounts for cromolyn’s formerly widespread use in children, especially during ages of rapid growth. Its place in treatment of childhood asthma has lately diminished, however, because of the significantly greater efficacy of even low-dose corticosteroid treatment and because of the availability of an alternate nonsteroidal controller class of medication, the leukotriene pathway inhibitors (see below).

LEUKOTRIENE PATHWAY INHIBITORS Because of the evidence of leukotriene involvement in many inflammatory diseases (see Chapter 18) and in anaphylaxis, considerable effort has been expended on the development of drugs that block their synthesis or interaction with their receptors. Leukotrienes result from the action of 5-lipoxygenase on arachidonic acid and are synthesized by a variety of inflammatory cells in the airways, including eosinophils, mast cells, macrophages, and basophils. Leukotriene B4 (LTB4 ) is a potent neutrophil chemoattractant, and LTC 4 and LTD 4 exert many effects known to occur in asthma, including bronchoconstriction, increased bronchial reactivity, mucosal edema, and mucus hypersecretion. Antigen challenge of sensitized human lung tissue in vitro results in the generation of leukotrienes. Inhalation of leukotrienes by volunteers with asthma results not only in bronchoconstriction but also in an increase in bronchial reactivity. Two approaches to interrupting the leukotriene pathway have been pursued: inhibition of 5-lipoxygenase, thereby preventing leukotriene synthesis; and inhibition of the binding of LTD 4 to its receptor on target tissues, thereby preventing its action. Efficacy in blocking airway responses to exercise and to antigen challenge has been shown for drugs in both categories: zileuton, a 5-lipoxygenase inhibitor, and zafirlukast and montelukast, LTD4 -receptor antagonists (Figure 20–6). All have been shown to improve asthma control and to reduce the frequency of asthma exacerbations in clinical trials. Their effects on symptoms, airway caliber, bronchial reactivity, and airway inflammation are less marked than the effects of ICS, but they are more nearly equal in reducing the frequency of exacerbations. Their principal advantage is that they are taken orally; some patients—especially children—comply poorly with inhaled therapies. Montelukast is approved for children as young as 12 months.


FIGURE 20–6 Structures of leukotriene receptor antagonists (montelukast, zafirlukast) and of the 5-lipoxygenase inhibitor (zileuton). Some patients appear to have particularly favorable responses, but no clinical features aside from the subclass of patients with aspirin-sensitive asthma described below allow identification of “responders” before a trial of therapy. In the USA, zileuton is approved for use in an oral dosage of 1200 mg of the sustained-release form twice daily; zafirlukast, 20 mg twice daily; and montelukast, 10 mg (for adults) or 4 mg (for children) once daily. Trials with leukotriene inhibitors have demonstrated an important role for leukotrienes in aspirin-induced asthma. It has long been known that in 5–10% of patients with asthma, ingestion of even a very small dose of aspirin causes profound bronchoconstriction and symptoms of systemic release of histamine, such as flushing and abdominal cramping. Because this reaction to aspirin is not associated with any evidence of allergic sensitization to aspirin or its metabolites and because it is produced by any of the nonsteroidal antiinflammatory agents, it is thought to result from inhibition of prostaglandin synthetase (cyclooxygenase), shifting arachidonic acid metabolism from the prostaglandin to the leukotriene pathway, especially in platelets adherent to circulating neutrophils. Support for this idea was provided by the demonstration that leukotriene pathway inhibitors impressively reduce the response to aspirin challenge and improve overall control of asthma on a day-to-day basis. Of these agents, montelukast is by far the most prescribed, probably because it can be taken without regard to meals, because of the convenience of once-daily treatment, and because of patient fear of inhaled corticosteroids. Zileuton is the least prescribed because of reports of liver toxicity. The receptor antagonists appear to have little toxicity. Early reports of Churg-Strauss syndrome (a systemic vasculitis accompanied by worsening asthma, pulmonary infiltrates, and eosinophilia) appear to have been coincidental, with the syndrome unmasked by the reduction in prednisone dosage made possible by the addition of zafirlukast or montelukast.

OTHER DRUGS IN THE TREATMENT OF ASTHMA Anti-IgE Monoclonal Antibodies


The development of a monoclonal antibody that targets IgE antibody itself was a novel approach to the treatment of asthma. The monoclonal antibody-developed omalizumab was raised in mice and then “humanized,” making it less likely to cause sensitization when given to human subjects. Because its specific target is the portion of IgE that binds to its receptors (Fcε-R1 and Fcε-R2 receptors) on mast cells and other inflammatory cells, omalizumab inhibits the binding of IgE but does not activate IgE already bound to mast cells and thus does not provoke mast cell degranulation. Omalizumab’s use is restricted to patients with evidence of allergic sensitization, and the dose administered is adjusted for total IgE level and body weight. Given by subcutaneous injection every 2–4 weeks to asthmatic patients, it lowers free plasma IgE to undetectable levels and significantly reduces the magnitude of both early and late bronchospastic responses to antigen challenge. Omalizumab’s most important clinical effect is reduction in the frequency and severity of asthma exacerbations, even while enabling a reduction in corticosteroid requirements. It also lessens asthma severity and improves coincident nasal and conjunctival symptoms of allergic rhinitis. Combined analysis of several clinical trials has shown that the patients most likely to respond are those with a history of repeated exacerbations, a high requirement for corticosteroid treatment, and poor pulmonary function. Similarly, the exacerbations most prevented are the ones most important to prevent: omalizumab treatment reduced exacerbations requiring hospitalization by 88%. These benefits justify the high cost of this treatment in selected individuals with severe disease characterized by frequent exacerbations. The addition of omalizumab to standard, guidelines-based therapy for asthmatic inner-city children and adolescents has been shown to significantly improve overall asthma control, reduce the need for other medications, and nearly eliminate the seasonal peaks in exacerbations attributed to viral respiratory infections. This last, unexpected, finding will likely encourage further development of IgEtargeted therapies. There is also evidence of effectiveness of omalizumab treatment for chronic urticaria (for which the drug is now approved) and peanut allergy.

FUTURE DIRECTIONS OF ASTHMA THERAPY Ironically, the effectiveness of ICS as a treatment for most patients with asthma, especially for young adults with allergic asthma, may have retarded recognition that the term “asthma” encompasses a heterogeneous collection of disorders, many of which are poorly responsive to corticosteroid treatment. The existence of different forms or subtypes of asthma has actually long been recognized, as is implied by the use of modifying terms such as “extrinsic” versus “intrinsic,” “aspirin-sensitive,” “adult onset,” “steroid-dependent,” “exacerbation-prone,” “seasonal,” “post-viral,” and “obesity-related” to describe asthma in particular patients. More rigorous description of asthma phenotypes, based on cluster analysis of multiple clinical, physiological, and laboratory features, including analysis of blood and sputum inflammatory cell assessments, has identified as many as five different asthma phenotypes. The key question raised by this approach is whether the phenotypes respond differently to available asthma treatments. The most persuasive evidence of the existence of different asthma phenotypes is the demonstration of differences in the pattern of gene expression in the airway epithelium among asthmatic and healthy subjects (Figure 20–7). Compared with healthy controls, half of the asthmatic participants overexpressed three genes up-regulated in airway epithelial cells by IL-13, a signature cytokine of TH2 lymphocytes. These genes express the proteins periostin, CLCA1, and serpinB2. The other half of the population did not, with some (but not all) having a pattern of airway epithelial cell gene expression suggesting exposure to IL-17. These findings suggest that fundamentally different pathophysiologic mechanisms may underlie the clinical expression of asthma even among patients with mild forms of the disease. The participants with overexpression of genes up-regulated by IL-13 are referred to as having a “TH2 molecular phenotype” (or “endotype”) of asthma. The other subjects, who did not overexpress these genes, are described as having a “non-TH2 molecular phenotype.” The TH2-type asthmatic subjects on average had more sputum and blood eosinophilia, positive skin test results, higher levels of IgE, and greater expression of certain mucin genes, but there was overlap between the groups. Though subjects in both groups showed improvement in their FEV1 after treatment with albuterol, their response to treatment with 6 weeks of ICS was quite different; FEV1 improved only in the TH2-type subjects. If these findings are valid—and they have held up well so far—the implications are farreaching; they would mean that many, perhaps as many as half of, patients with mild-moderate asthma do not respond to inhaled corticosteroid therapy. The proportion of non-inhaled corticosteroid responders among severe “steroid-resistant” asthma could be much higher.


FIGURE 20–7 Cluster analysis of subjects according to their expression of periostin, chloride channel regulator 1 (CLCA1), and serpinB2 in bronchial epithelium. Note that cluster 1, including all subjects with high expression of these genes, contains only asthmatic subjects (A; n = 22). These are referred to as having TH2-high asthma, because the three genes are known to be up-regulated in epithelial cells by IL-13, a prototypic TH2-cytokine. Cluster 2 includes all subjects with lower levels of expression and contains all healthy control subjects (H; n = 28) and approximately half of the subjects with asthma (n = 20) now referred to as having TH2-low asthma. (B) Responsiveness of TH2-high vs TH2-low asthmatic subjects to inhaled steroids and to placebo in a randomized controlled trial. FEV1 measured at baseline (week 0), after 4 and 8 weeks on daily fluticasone (500 mcg twice daily), and 1 week after the cessation of


fluticasone (week 9). (Reproduced, with permission, of the American Thoracic Society. Copyright © 2014 American Thoracic Society. Woodruff PG et al: T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med 2009;180:388. Official Journal of the American Thoracic Society.) Current research has focused on further exploring molecular phenotypes in asthma and in finding effective treatments for each group. An investigational IL-13 receptor antagonist, lebrikizumab, was tested in patients with moderately severe asthma. Though its effects fell short of significance in the study as a whole, when investigators stratified the subjects based on serum levels of periostin (one of the genes up-regulated in the “TH2 molecular phenotype”), the drug was found to be effective in participants with high levels of periostin but not in those with lower levels. A multicenter trial is embarking on a prospective double-blind, placebo-controlled trial of ICS versus tiotropium in asthmatic subjects characterized as TH2 or non-TH2 by analysis of their induced sputum samples for eosinophil number and for expression of TH2dependent genes, with the hope of identifying patients that are optimally treated by one or the other medications. The pace of advance in the scientific description of the immunopathogenesis of asthma has spurred the development of many new therapies that target different sites in the immune cascade. These include monoclonal antibodies directed against cytokines (IL-4, IL-5, IL-13), antagonists of cell adhesion molecules, protease inhibitors, and immunomodulators aimed at shifting CD4 lymphocytes from the TH2 to the TH1 phenotype or at selective inhibition of the subset of TH2 lymphocytes directed against particular antigens. As with the development of the IL-13 receptor antagonist, the identification of subgroups of asthma that are most likely to benefit from therapy may finally herald the advent of truly personalized asthma therapy.

CLINICAL PHARMACOLOGY OF DRUGS USED IN THE TREATMENT OF ASTHMA Asthma is best thought of as a disease in two time domains. In the present domain, it is important for the distress it causes—cough, nocturnal awakenings, and shortness of breath that interferes with the ability to exercise or to pursue desired activities. For mild asthma, occasional inhalation of a bronchodilator may be all that is needed. For more severe asthma, treatment with a long-term controller, like an inhaled corticosteroid, is necessary to relieve symptoms and restore function. The second domain of asthma is the risk it presents of future events, such as exacerbations, or of progressive loss of pulmonary function. Satisfaction with the ability to control symptoms and maintain function by frequent use of an inhaled β2 agonist does not mean that the risk of future events is also controlled. In fact, use of two or more canisters of an inhaled β agonist per month is a marker of increased risk of asthma fatality. The challenges of assessing severity and adjusting therapy for these two domains of asthma are different. For relief of distress in the present domain, the key information is obtained by asking specific questions about the frequency and severity of symptoms, the frequency of rescue use of an inhaled β2 agonist, the frequency of nocturnal awakenings, and the ability to exercise. The best predictor of the risk for future exacerbations is the frequency of their occurrence in the past. Without such a history, estimation of risk is more difficult. In general, patients with poorly controlled symptoms have a heightened risk of exacerbations in the future, but some patients seem unaware of the severity of their airflow obstruction (sometimes described as “poor perceivers”) and can be identified only by measurement of pulmonary function, as by spirometry. Reductions in the FEV 1 correlate with heightened risk of future attacks of asthma. Other possible markers of heightened risk are unstable pulmonary function (large variations in FEV1 from visit to visit, large change with bronchodilator treatment), extreme bronchial reactivity, or high numbers of eosinophils in sputum or of nitric oxide in exhaled air. Assessment of these features may identify patients who need increases in therapy for protection against exacerbations.

BRONCHODILATORS Bronchodilators, such as inhaled albuterol, are rapidly effective, safe, and inexpensive. Patients with only occasional symptoms of asthma require no more than an inhaled bronchodilator taken on an as-needed basis. If symptoms require this “rescue” therapy more than twice a week, if nocturnal symptoms occur more than twice a month, or if the FEV1 is less than 80% predicted, additional treatment is needed. The treatment first recommended is a low dose of an inhaled corticosteroid, although treatment with a leukotriene receptor antagonist or with cromolyn may be used. Theophylline is now largely reserved for patients in whom symptoms remain poorly controlled despite the combination of regular treatment with an inhaled anti-inflammatory agent and as-needed use of a β2 agonist. If the addition of theophylline fails to improve symptoms or if adverse effects become bothersome, it is important to check the plasma level of theophylline to be sure it is in the therapeutic range (10–20 mg/L). An important caveat for patients with mild asthma is that although the risk of a severe, life-threatening attack is lower than in patients with severe asthma, it is not zero. All patients with asthma should be instructed in a simple action plan for severe, frightening attacks: to take up to four puffs of albuterol every 20 minutes over 1 hour. If they do not note clear improvement after the first four puffs, they should take the additional treatments while on their way to an emergency department or other higher level of care.


MUSCARINIC ANTAGONISTS Inhaled muscarinic antagonists have so far earned a limited place in the treatment of asthma. The effects of short-acting agents (eg, ipratropium bromide) on baseline airway resistance is nearly as great as, but no greater than, those of the sympathomimetic drugs, so they are used largely as alternative therapies for patients intolerant of β2 -adrenoceptor agonists. The airway effects of antimuscarinic and sympathomimetic drugs given in full doses have been shown to be additive only in patients with severe airflow obstruction who present for emergency care. The long-acting antimuscarinic agents tiotropium and aclidinium have not yet earned a place in the treatment for asthma, although tiotropium has been shown to be as effective as a long-acting β2 agonist when used in addition to an inhaled corticosteroid. As a treatment for COPD, these agents improve functional capacity, presumably through their action as bronchodilators, and reduce the frequency of exacerbations, through mechanisms not yet defined. Although it was predicted that muscarinic antagonists would dry airway secretions and interfere with mucociliary clearance, direct measurements of fluid volume secretion from airway submucosal glands in animals show that atropine decreases baseline secretory rates only slightly. The drugs do, however, inhibit the increase in mucus secretion caused by vagal stimulation. No cases of inspissation of mucus have been reported following administration of these drugs.

CORTICOSTEROIDS If asthmatic symptoms occur frequently, or if significant airflow obstruction persists despite bronchodilator therapy, inhaled corticosteroids should be started. For patients with severe symptoms or severe airflow obstruction (eg, FEV1 < 50% predicted), initial treatment with a combination of inhaled and oral corticosteroid (eg, 30 mg/d of prednisone for 10 days) is appropriate. Once clinical improvement is noted, usually after 7–10 days, the ICS should be continued, but the oral dose should be tapered to the minimum necessary to control symptoms. An issue for inhaled corticosteroid treatment is patient adherence. Analysis of prescription renewals shows that only a minority of patients take corticosteroids regularly. This may be a function of a general “steroid phobia” fostered by emphasis in the lay press on the hazards of long-term oral corticosteroid therapy and by ignorance of the difference between corticosteroids and anabolic steroids, taken to enhance muscle strength by now-infamous athletes. This fear of corticosteroid toxicity makes it hard to persuade patients whose symptoms have improved after starting treatment that they should continue it for protection against attacks. This context accounts for the interest in reports that instructing patients with mild but persistent asthma to take inhaled corticosteroid therapy only when their symptoms worsen is as effective in maintaining pulmonary function and preventing attacks as is taking the inhaled corticosteroid twice each day (see above). In patients with more severe asthma whose symptoms are inadequately controlled by a standard dose of an inhaled corticosteroid, two options may be considered: to double the dose of inhaled corticosteroid or to combine it with another drug. The addition of theophylline or a leukotriene-receptor antagonist modestly increases asthma control, but the most impressive benefits are afforded by addition of a long-acting inhaled β2 -receptor agonist (LABA, eg, salmeterol or formoterol). Many studies have shown this combination therapy to be more effective than doubling the dose of the inhaled corticosteroid for improving asthma control. Combinations of an inhaled corticosteroid and a LABA in a single inhaler are now commonly available in fixed-dose combinations (eg, fluticasone and salmeterol [Advair]; budesonide and formoterol [Symbicort]; and mometasone and formoterol [Dulera]). The rapid onset of action of formoterol enables novel use of the combination of an inhaled corticosteroid with this long-acting β agonist. Several studies have confirmed that twice-daily plus as-needed inhalation of budesonide and formoterol is as effective in preventing asthma exacerbations as twice-daily inhalation of a four-times-higher dose of budesonide with only albuterol for relief of symptoms. Use of this flexible dosing strategy is widespread in Europe but is not approved in the USA. Offsetting the clear benefits is evidence of a statistically significant increase in the very low risk of fatal or near-fatal asthma attacks from use of a long-acting β agonist, perhaps even when taken in combination with an inhaled corticosteroid. This evidence prompted the FDA to issue a “black box” warning of this risk, especially in African Americans. The FDA did not withdraw approval of these drugs, for it recognizes that they are clinically effective. The major implications of the black box warning for the practitioner are that: (1) patients with mild to moderate asthma should be treated with a low-dose inhaled corticosteroid alone, and additional therapy considered only if their asthma is not well controlled; and, (2) if their asthma is not well controlled, the possible increase in risk of a rare event, asthma fatality, should be discussed in presenting the options for treatment—an increase to a higher dose of the inhaled corticosteroid versus addition of a long-acting β agonist. The FDA’s warning has not so far had much effect on prescriptions for inhaled corticosteroid/long-acting β-agonist combinations, probably because their combination in a single inhaler offers several advantages. Combination inhalers are convenient; they ensure that the long-acting β agonist will not be taken as monotherapy (known not to protect against attacks); and they produce prompt, sustained improvements in clinical symptoms and pulmonary function and reduce the frequency of exacerbations. In patients prescribed such combination treatment, it is important to provide explicit instructions that a rapid-acting inhaled β2 agonist, such as albuterol, should still be used as needed for relief of acute symptoms.


LEUKOTRIENE ANTAGONISTS; CROMOLYN & NEDOCROMIL A leukotriene receptor antagonist taken as an oral tablet is an alternative to inhaled corticosteroid treatment in patients with symptoms occurring more than twice a week or those who are awakened from sleep by asthma more than twice a month. This place in asthma therapy was once held by cromolyn and nedocromil, but neither is now available in the USA. Although these treatments are not as effective as even a low dose of an inhaled corticosteroid, both avoid the issue of “steroid phobia” described above and are commonly used in the treatment of children. The leukotriene receptor antagonist montelukast (Singulair) is widely prescribed, especially by primary care providers. This drug, taken orally, is easy to administer and appears to be used more regularly than ICS. Leukotriene receptor antagonists are rarely associated with troublesome side effects. Because of concerns over the possible long-term toxicity of systemic absorption of ICS, this maintenance therapy is widely used for treating children in the USA, particularly those who have concurrent symptomatic allergic rhinitis, which is also effectively treated by montelukast.

ANTI-IGE MONOCLONAL ANTIBODY Treatment with omalizumab, the monoclonal humanized anti-IgE antibody, is reserved for patients with chronic severe asthma inadequately controlled by high-dose inhaled corticosteroid plus long-acting β-agonist combination treatment. Omalizumab reduces lymphocytic, eosinophilic bronchial inflammation, oral and inhaled corticosteroid dose requirements, and the frequency and severity of exacerbations. It is reserved for patients with demonstrated IgE-mediated sensitivity (by positive skin test or radioallergosorbent test [RAST] to common allergens) and an IgE level within a range that can be reduced sufficiently by twice-weekly subcutaneous injection. In addition to its high cost, several factors have limited the use of omalizumab. First, it must be given as a subcutaneous injection every 2–4 weeks. Although the antibody has been humanized, it nonetheless can cause anaphylactic reactions in 0.1–0.2% of patients taking the drug. For this reason, it cannot be self-administered but must be given in a physician’s office or infusion center equipped to manage an anaphylactic reaction. Furthermore, patients receiving omalizumab must be monitored for a period of time after the injection. Even then, anaphylactic reactions have been reported over 24 hours after the injection, even in patients who had safely received the drug before. Finally, in clinical trials, a slight excess of malignancies was observed in patients receiving omalizumab compared with those assigned to the placebo group.

OTHER ANTI-INFLAMMATORY THERAPIES For the 5–10% of the asthmatic population with severe asthma inadequately controlled by standard therapies, including high-dose inhaled corticosteroid treatment, the development of an alternative treatment is an important unmet medical need. The initial promise of oral methotrexate or gold salt injections has not been fulfilled. While the benefit from treatment with cyclosporine seems real, this drug’s toxicity makes this finding only a source of hope that other immunomodulatory therapies will ultimately emerge. Advances in understanding the immunopathogenesis of asthma may permit the identification of specific phenotypes of asthma and identification of biomarkers of their importance in particular patients. In this respect, asthma may benefit from the rapid advances in treatments developed for other chronic inflammatory conditions such as rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease.

MANAGEMENT OF ACUTE ASTHMA The treatment of acute attacks of asthma in patients reporting to the hospital requires close, continuous clinical assessment and repeated objective measurement of lung function. For patients with mild attacks, inhalation of a β2 -receptor agonist is as effective as subcutaneous injection of epinephrine. Both of these treatments are more effective than intravenous administration of aminophylline (a soluble salt of theophylline). Severe attacks require treatment with oxygen, frequent or continuous administration of aerosolized albuterol, and systemic treatment with prednisone or methylprednisolone (0.5 mg/kg every 6–12 hours). Even this aggressive treatment is not invariably effective, and patients must be watched closely for signs of deterioration. General anesthesia, intubation, and mechanical ventilation of asthmatic patients cannot be undertaken lightly but may be lifesaving if respiratory failure supervenes.

PROSPECTS FOR PREVENTION The high prevalence of asthma in the developed world and its rapid increases in the developing world call for a strategy for primary prevention. Strict antigen avoidance during infancy, once thought to be sensible, has now been shown to be ineffective. In fact, growing up from birth on a farm with domestic animals or in a household where cats or dogs are kept as pets appears to protect against developing asthma. The best hope seems to lie in understanding the mechanisms by which microbial exposures during infancy foster development of a balanced immune response and then mimicking the effects of natural environmental exposures through administration


of harmless microbial commensals (probiotics) or of nutrients that foster their growth (prebiotics) in the intestinal tract during the critical period of immune development in early infancy.

TREATMENT OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE (COPD) COPD is characterized by airflow limitation that is not fully reversible with bronchodilator treatment. The airflow limitation is usually progressive and is believed to reflect an abnormal inflammatory response of the lung to noxious particles or gases. The condition is most often a consequence of prolonged habitual cigarette smoking, but approximately 15% of cases occur in nonsmokers. Although COPD is different from asthma, some of the same drugs are used in its treatment. This section discusses the drugs that are useful in both conditions. Although asthma and COPD are both characterized by airway inflammation, reduction in maximum expiratory flow, and episodic exacerbations of airflow obstruction, most often triggered by viral respiratory infection, they differ in many important respects. Most important among them are differences in the populations affected, characteristics of airway inflammation, reversibility of airflow obstruction, responsiveness to corticosteroid treatment, and course and prognosis. Compared to asthma, COPD occurs in older patients, is associated with neutrophilic rather than eosinophilic inflammation, is poorly responsive even to high-dose inhaled corticosteroid therapy, and is associated with progressive, inexorable loss of pulmonary function over time, especially with continued cigarette smoking. Despite these differences, the approaches to treatment are similar, although the benefits expected (and achieved) are less for COPD than for asthma. For relief of acute symptoms, inhalation of a short-acting β agonist (eg, albuterol), of an anticholinergic drug (eg, ipratropium bromide), or of the two in combination is usually effective. For patients with persistent symptoms of exertional dyspnea and limitation of activities, regular use of a long-acting bronchodilator, whether a long-acting β agonist such as salmeterol or a long-acting anticholinergic (eg, tiotropium) is indicated. For patients with severe airflow obstruction or with a history of prior exacerbations, regular use of an inhaled corticosteroid reduces the frequency of exacerbations. Theophylline may have a particular place in the treatment of COPD, as it may improve contractile function of the diaphragm, thus improving ventilatory capacity. The major difference in treatment of these conditions centers on management of exacerbations. The use of antibiotics in this context is routine in COPD, because such exacerbations involve bacterial infection of the lower airways far more often in COPD than in asthma.

SUMMARY Drugs Used in Asthma



PREPARATIONS AVAILABLE


REFERENCES Pathophysiology of Airway Disease


Holgate ST : Pathophysiology of asthma: What has our current understanding taught us about new therapeutic approaches? J Allergy Clin Immunol 2011;128:495. Locksley RM: Asthma and allergic inflammation. Cell 2010;140:777. Lotvall J et al: Asthma endotypes: A new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol 2011;127:355. Martinez FD, Vercelli D: Asthma. Lancet 2013;382:1360.

Asthma Treatment Bateman ED et al: Overall asthma control: T he relationship between current control and future risk. J Allergy Clin Immunol 2010;125:600. Bel EH: Mild asthma. N Engl J Med 2013;369:2362. Busse WW: Asthma diagnosis and treatment: Filling in the information gaps. J Allergy Clin Immunol 2011;128:740. National Heart, Lung, and Blood Institute, National Asthma Education and Prevention Program. Expert Panel Report 3: Guidelines for the diagnosis and management of asthma. National Heart, Lung, and Blood Institute; Revised August 2007. NIH publication no. 07-4051. http://www.nhlbi.nih.gov/guidelines/asthma/asthgdln.pdf. von Mutius E, Drazen JM: A patient with asthma seeks medical advice in 1828, 1928, and 2012. N Engl J Med 2012;366:827.

Beta Agonists Ducharme FM et al: Addition of long-acting beta2-agonists to inhaled corticosteroids versus same dose inhaled corticosteroids for chronic asthma in adults and children. Cochrane Database Syst Rev 2010;(5):CD005535. Ducharme FM et al: Addition of long-acting beta2-agonists to inhaled steroids versus higher dose inhaled steroids in adults and children with persistent asthma. Cochrane Database Syst Rev 2010:CD005533. Papi A et al: Beclometasone-formoterol as maintenance and reliever treatment in patients with asthma: a double-blind, randomised controlled trial. Lancet Respir Med 2013;1:23.

Methylxanthines & Roflumilast Barnes PJ: T heophylline. Am J Respir Crit Care Med 2013;188:901. Beghe B, Rabe KF, Fabbri LM: Phosphodiesterase-4 inhibitor therapy for lung diseases. Am J Respir Crit Care Med 2013;188:271. Rabe KF: Roflumilast for the treatment of chronic obstructive pulmonary disease. Expert Rev Respir Med 2010;4:543.

Cromolyn & Nedocromil Guevara J et al: Inhaled corticosteroids versus sodium cromoglycate in children and adults with asthma. Cochrane Database Syst Rev 2006;(2):CD003558. Yoshihara S et al: Effects of early intervention with inhaled sodium cromoglycate in childhood asthma. Lung 2006;184:63.

Corticosteroids Barnes P: How corticosteroids control inflammation: Quintiles Prize Lecture 2005. Br J Pharmacol 2006;148:245. Beasley R et al: Combination corticosteroid/beta-agonist inhaler as reliever therapy: A solution for intermittent and mild asthma? J Allergy Clin Immunol 2014;133:39. Boushey HA et al: Daily versus as-needed corticosteroids for mild persistent asthma. N Engl J Med 2005;352:1519. Suissa S et al: Low-dose inhaled corticosteroids and the prevention of death from asthma. N Engl J Med 2000;343:332.

Antimuscarinic Drugs Gross N: Anticholinergic agents in asthma and COPD. Eur J Pharmacol 2006;8:533. Lee AM, Jacoby DB, Fryer AD: Selective muscarinic receptor antagonists for airway diseases. Curr Opin Pharmacol 2001;1:223. Peters SP et al: T iotropium bromide step-up therapy for adults with uncontrolled asthma. N Engl J Med 2010;363:1715.

Leukotriene Pathway Inhibitors Calhoun WJ: Anti-leukotrienes for asthma. Curr Opin Pharmacol 2001;1:230. Laidlaw T M et al: Cysteinyl leukotriene overproduction in aspirin-exacerbated respiratory disease is driven by platelet-adherent leukocytes. Blood 2012;119:3790. Wang L et al: Cost-effectiveness analysis of fluticasone versus montelukast in children with mild-to-moderate persistent asthma in the Pediatric Asthma Controller T rial. J Allergy Clin Immunol 2011;127:161.

Anti-IgE Therapy Busse WW et al: Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N Engl J Med 2011;364:1005. Walker S et al: Anti-IgE for chronic asthma in adults and children. Cochrane Database Syst Rev 2006;(2):CD003559.

Future Directions of Asthma Therapy


Chang T S et al: Childhood asthma clusters and response to therapy in clinical trials. J Allergy Clin Immunol 2014;133:363. Corren J et al: Lebrikizumab treatment in adults with asthma. N Engl J Med 2011;365:1088. Haldar P et al: Cluster analysis and clinical asthma phenotypes. Am J Respir Crit Care Med 2008;178:218. Lotvall J et al: Asthma endotypes: A new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol 2011;127:355. Moore WC et al: Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med 2010;181:315. Woodruff PG et al: T -helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med 2009;180:388.

Management of Acute Asthma Lazarus SC: Clinical practice. Emergency treatment of asthma. N Engl J Med 2010;363:755.

Prospects for Prevention Martinez FD: New insights into the natural history of asthma: Primary prevention on the horizon. J Allergy Clin Immunol 2011;128:939.

Treatment of COPD Global Initiative for Chronic Obstructive Lung Disease, Inc: In: Global Strategy for Diagnosis, Management, and Prevention of COPD, 2014. http://www.goldcopd.org/uploads/users/files/GOLD_Report2014_Feb07.pdf. Niewoehner DE: Clinical practice. Outpatient management of severe COPD. N Engl J Med 2010;362:1407. Matera MG, Page CP, Cazzola M: Novel bronchodilators for the treatment of chronic obstructive pulmonary disease. T rends Pharmacol Sci 2011;32:495. Vestbo J et al: Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med 2013;187:347. Vogelmeier C et al: T iotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med 2011;364;1093.

CASE STUDY ANSWER This patient demonstrates the destabilizing effects of a respiratory infection on asthma, and the parents demonstrate the common (and dangerous) phobia about “overuse” of bronchodilator or steroid inhalers. The patient has signs of imminent respiratory failure, including her refusal to lie down, her fear, and her tachycardia—which cannot be attributed to her minimal treatment with albuterol. Critically important immediate steps are to administer high-flow oxygen and to start albuterol by nebulization. Adding ipratropium (Atrovent) to the nebulized solution is recommended. A corticosteroid (0.5–1.0 mg/kg of methylprednisolone) should be administered intravenously. It is also advisable to alert the intensive care unit, because a patient with severe bronchospasm who tires can slip into respiratory failure quickly, and intubation can be difficult. Fortunately, most patients treated in hospital emergency departments do well. Asthma mortality is rare (fewer than 5000 deaths per year among a population of 20 million asthmatics in the USA), and when it occurs, it is often out of hospital. Presuming this patient recovers, she needs adjustments to her therapy before discharge. The strongest predictor of severe attacks of asthma is their occurrence in the past. Thus, this patient needs to be started on a long-term controller, especially an inhaled corticosteroid, and needs instruction in an action plan for managing severe symptoms. This can be as simple as advising her and her parents that if she has a severe attack that frightens her, she can take up to four puffs of albuterol every 15 minutes, but if the first treatment does not bring significant relief, she should take the next four puffs while on her way to an emergency department or urgent care clinic. She should also be given a prescription for prednisone, with instructions to take 40–60 mg orally for severe attacks, but not to wait for it to take effect if she remains severely short of breath even after albuterol inhalations. Asthma is a chronic disease, and good care requires close follow-up and creation of a provider-patient partnership for optimal management.


SECTION V DRUGS THAT ACT IN THE CENTRAL NERVOUS SYSTEM


CHAPTER

21 Introduction to the Pharmacology of CNS Drugs John A. Gray, MD, PhD, & Roger A. Nicoll, MD

Drugs acting in the central nervous system (CNS) were among the first to be discovered by primitive humans and are still the most widely used group of pharmacologic agents. These include medications used to treat a wide range of neurologic and psychiatric conditions as well as drugs that relieve pain, suppress nausea, and reduce fever, among other symptoms. In addition, many CNS-acting drugs are used without prescription to increase the sense of well-being. Due to their complexity, the mechanisms by which various drugs act in the CNS have not always been clearly understood. In recent decades, however, dramatic advances have been made in the methodology of CNS pharmacology. It is now possible to study the action of a drug on individual neurons and even single receptors within synapses. The information obtained from such studies is the basis for several major developments in studies of the CNS. First, it is clear that nearly all drugs with CNS effects act on specific receptors that modulate synaptic transmission. While a few agents such as general anesthetics and alcohol may have nonspecific actions on membranes (although these exceptions are not fully accepted), even these non-receptor-mediated actions result in demonstrable alterations in synaptic transmission. Second, drugs are among the most valuable tools for studying CNS function, from understanding the mechanism of convulsions to the laying down of long-term memory. Both agonists that mimic natural transmitters (and in many cases that are more selective than the endogenous substances) and antagonists are extremely useful in such studies. Third, unraveling the actions of drugs with known clinical efficacy has led to some of the most fruitful hypotheses regarding the mechanisms of disease. For example, information about the action of antipsychotic drugs on dopamine receptors has provided the basis for important hypotheses regarding the pathophysiology of schizophrenia. Studies of the effects of a variety of agonists and antagonists on γ-aminobutyric acid (GABA) receptors have resulted in new concepts pertaining to the pathophysiology of several diseases, including anxiety and epilepsy. A full appreciation of the effects of a drug on the CNS requires an understanding of the multiple levels of brain organization, from genes to circuits to behavior. This chapter provides an introduction to the functional organization of the CNS and its synaptic transmitters as a basis for understanding the actions of the drugs described in the following chapters.

ORGANIZATION OF THE CNS The CNS comprises the brain and spinal cord and is responsible for integrating sensory information and generating motor output and other behaviors needed to successfully interact with the environment and enhance species survival. The human brain contains about 100 billion interconnected neurons surrounded by various supporting glial cells. Throughout the CNS, neurons are either clustered into groups called nuclei or are present in layered structures such as the cerebellum or hippocampus. Connections among neurons both within and between these clusters form the circuitry that regulates information flow through the CNS.

Neurons Neurons are electrically excitable cells that process and transmit information via an electrochemical process. There are many types of neurons in the CNS, and they are classified in the following ways: by function, by location, and by the neurotransmitter they release. The typical neuron possesses a cell body (or soma) and specialized processes called dendrites and axons (Figure 21–1). Dendrites, which form highly branched complex dendritic “trees,” receive and integrate the input from other neurons and conduct this information to the cell body. The axon carries the output signal of a neuron from the cell body, sometimes over long distances. Neurons may have hundreds of dendrites but generally have only one axon, though axons may branch distally to contact multiple targets. The axon terminal makes contact with other neurons at specialized junctions, called synapses, where neurotransmitter chemicals are released that interact with receptors on other neurons.


FIGURE 21–1 Neurons and glia in the CNS. A typical neuron has a cell body (or soma) that receives the synaptic responses from the dendritic tree. These synaptic responses are integrated at the axon initial segment, which has a high concentration of voltage-gated sodium channels. If an action potential is initiated, it propagates down the axon to the synaptic terminals, which contact other neurons. The axon of long-range projection neurons is insulated by a myelin sheath derived from specialized membrane processes of oligodendrocytes, analogous to the Schwann cells in the peripheral nervous system. Astrocytes perform supportive roles in the CNS, and their processes are closely associated with neuronal synapses. (see Figures 21-4 and 21-7).

Neuroglia In addition to neurons, there are a large number of non-neuronal support cells, called neuroglia or glia, that perform a variety of essential functions in the CNS. Astrocytes are the most abundant glial cells in the brain and play homeostatic support roles, including providing metabolic nutrients to neurons and maintaining extracellular ion concentrations. In addition, astrocyte processes are closely associated with neuronal synapses where they are involved in the removal and recycling of neurotransmitters after release and play increasingly appreciated roles in regulating neurotransmission (see below). Oligodendrocytes are cells that wrap around the axons of projection neurons in the CNS forming the myelin sheath (Figure 22–1). Similar to the Schwann cells in peripheral neurons, the myelin sheath created by the oligodendrocytes insulates the axons and increases the speed of signal propagation. Damage to oligodendrocytes occurs in multiple sclerosis and thus is a target of drug discovery efforts. Microglia are specialized macrophages derived from the bone marrow that are found in the CNS and are the major immune defense system in the brain. These cells are actively involved in neuroinflammatory processes in many pathological states including neurodegenerative diseases.

Blood-Brain Barrier The blood-brain barrier (BBB) is a protective functional separation of the circulating blood from the extracellular fluid of the CNS that limits the penetration of substances, including drugs. This separation is accomplished by the presence of tight junctions between the capillary endothelial cells as well as a surrounding layer of astrocyte end-feet. Therefore, to enter the CNS, drugs must either be highly hydrophobic or engage specific transport mechanisms. For example, the second-generation antihistamines cause less drowsiness because they were developed to be significantly more polar than older antihistamines, limiting their crossing of the BBB (see Chapter 16). Many nutrients, such as glucose and the essential amino acids, have specific transporters that allow them to cross the BBB. L-DOPA, a precursor of the neurotransmitter dopamine, can enter the brain using an amino acid transporter, whereas dopamine cannot cross the BBB. Thus, an orally administered drug, L-DOPA, but not dopamine, can be used to boost CNS dopamine levels in the treatment of Parkinson’s disease. Some parts of the brain, the so-called circumventricular organs, lack a normal BBB. These include regions that sample the blood, such as the area postrema vomiting center, and regions that secrete neurohormones into the circulation.


ION CHANNELS & NEUROTRANSMITTER RECEPTORS The membranes of neurons contain two types of channels defined on the basis of the mechanisms controlling their gating (opening and closing): voltage-gated and ligand-gated channels (Figure 21–2A and B). Voltage-gated channels respond to changes in the membrane potential of the cell. The voltage-gated sodium channel described in Chapter 14 for the heart is an example of this type of channel. In nerve cells, these channels are highly concentrated on the initial segment of the axon (Figure 21–1), which initiates the all-ornothing fast action potential and along the length of the axon where they propagate the action potential to the nerve terminal. There are also many types of voltage-sensitive calcium and potassium channels on the cell body, dendrites, and initial segment, which act on a much slower time scale and modulate the rate at which the neuron discharges. For example, some types of potassium channels opened by depolarization of the cell result in slowing of further depolarization and act as a brake to limit further action potential discharge. Plant and animal toxins that target various voltage-gated ion channels have been invaluable for studying the functions of these channels (see Box: Natural Toxins: Tools for Characterizing Ion Channels; Table 21–1). TABLE 21–1 Some toxins used to characterize ion channels.



FIGURE 21–2 Types of ion channels and neurotransmitter receptors in the CNS. A shows a voltage-gated channel in which a voltage sensor component of the protein controls the gating (broken arrow) of the channel. B shows a ligand-gated channel in which the binding of the neurotransmitter to the ionotropic channel receptor controls the gating (broken arrow) of the channel. C shows a G proteincoupled (metabotropic) receptor, which, when bound, activates a heterotrimeric G protein. D and E show two ways metabotropic receptors can regulate ion channels. The activated G protein can interact directly to modulate an ion channel (D) or the G protein can activate an enzyme that generates a diffusible second messenger (E), eg, cAMP, which can interact with the ion channel or can activate a kinase that phosphorylates and modulates a channel. Neurotransmitters exert their effects on neurons by binding to two distinct classes of receptor. The first class is referred to as ligand-gated channels, or ionotropic receptors. These receptors consist of multiple subunits, and binding of the neurotransmitter ligand directly opens the channel, which is an integral part of the receptor complex (see Figure 22–6). These channels are insensitive or


only weakly sensitive to membrane potential. Activation of these channels typically results in a brief (a few milliseconds to tens of milliseconds) opening of the channel. Ligand-gated channels are responsible for fast synaptic transmission typical of hierarchical pathways in the CNS (see following text).

Natural Toxins: Tools for Characterizing Ion Channels Evolution is tireless in the development of natural toxins. A vast number of variations are possible with even a small number of amino acids in peptides, and peptides make up only one of a broad array of toxic compounds. For example, the predatory marine snail genus Conus includes over 3000 different species. Each species kills or paralyzes its prey with a venom that contains 50–200 different peptides or proteins. Furthermore, there is little duplication of peptides among Conus species. Other animals with useful toxins include snakes, frogs, spiders, bees, wasps, and scorpions. Plant species with toxic (or therapeutic) substances are referred to in several other chapters of this book. Since many toxins act on ion channels, they provide a wealth of chemical tools for studying the function of these channels. In fact, much of our current understanding of the properties of ion channels comes from studies utilizing only a small percentage of the highly potent and selective toxins that are now available. The toxins typically target voltage-sensitive ion channels, but a number of very useful toxins block ionotropic neurotransmitter receptors. Table 21–1 lists some of the toxins most commonly used in research, their mode of action, and their source. The second class of neurotransmitter receptor is referred to as metabotropic receptors (Figure 21–2C). These are seventransmembrane G protein-coupled receptors of the type described in Chapter 2. The binding of neurotransmitter to this type of receptor does not result in the direct gating of a channel. Rather, binding to the receptor engages a G protein, which results in the production of second messengers that mediate intracellular signaling cascades such as those described in Chapter 2. In neurons, activation of metabotropic neurotransmitter receptors often leads to the modulation of voltage-gated channels. These interactions can occur entirely within the plane of the membrane and are referred to as membrane-delimited pathways (Figure 21– 2D). In this case, the G protein (often the βγ subunit) interacts directly with a voltage-gated ion channel. In general, two types of voltagegated ion channels are the targets of this type of signaling: calcium channels and potassium channels. When G proteins interact with calcium channels, they inhibit channel function. This mechanism accounts for the inhibition of neurotransmitter release that occurs when presynaptic metabotropic receptors are activated. In contrast, when these receptors are postsynaptic, they activate (cause the opening of) potassium channels, resulting in a slow postsynaptic inhibition. Metabotropic receptors can also modulate voltage-gated channels less directly by the generation of diffusible second messengers (Figure 21–2E). A classic example of this type of action is provided by the β adrenoceptor, which generates cAMP via the activation of adenylyl cyclase (see Chapter 2). Whereas membrane-delimited actions occur within microdomains in the membrane, second messenger-mediated effects can occur over considerable distances. Finally, an important consequence of the involvement of G proteins in receptor signaling is that, in contrast to the brief effect of ionotropic receptors, the effects of metabotropic receptor activation can last tens of seconds to minutes. Metabotropic receptors predominate in the diffuse neuronal systems in the CNS (see below).

THE SYNAPSE & SYNAPTIC POTENTIALS The communication between neurons in the CNS occurs through chemical synapses in the majority of cases. (A few instances of electrical coupling between neurons have been documented, and such coupling may play a role in synchronizing neuronal discharge. However, it is unlikely that these electrical synapses are an important site of drug action.) The events involved in synaptic transmission can be summarized as follows. An action potential propagating down the axon of the presynaptic neuron enters the synaptic terminal and activates voltage-sensitive calcium channels in the membrane of the terminal (see Figure 6–3). The calcium channels responsible for the release of neurotransmitter are generally resistant to the calcium channel-blocking agents discussed in Chapter 12 (verapamil, etc) but are sensitive to blockade by certain marine toxins and metal ions (see Tables 21–1 and 12–4). As calcium flows into the terminal, the increase in intraterminal calcium concentration promotes the fusion of synaptic vesicles with the presynaptic membrane. The neurotransmitter contained in the vesicles is released into the synaptic cleft and diffuses to the receptors on the postsynaptic membrane. The neurotransmitter binds to its receptor and opens channels (either directly or indirectly as described above) causing a brief change in membrane conductance (permeability to ions) of the postsynaptic cell. The time delay from the arrival of the presynaptic action potential to the onset of the postsynaptic response is approximately 0.5 ms. Most of this delay is consumed by the release process, particularly the time required for calcium channels to open.


FIGURE 21–3 Postsynaptic potentials and action potential generation. A (top) shows the voltage recorded upon entry of a microelectrode into a postsynaptic cell and subsequent recording of a resting membrane potential of −60 mV. Stimulation of an excitatory pathway (E1, left) generates transient depolarization called an excitatory postsynaptic potential (EPSP). Simultaneous activation of multiple excitatory synapses (E1 + E2, middle) increases the size of the depolarization, so that the threshold for action potential generation is reached. Alternatively, a train of stimuli from a single input can temporally summate to reach the threshold (E1 + E1, right). B (bottom) demonstrates the interaction of excitatory and inhibitory synapses. On the left, a suprathreshold excitatory stimulus (E3) evokes an action potential. In the center, an inhibitory pathway (I) generates a small hyperpolarizing current called an inhibitory postsynaptic potential (IPSP). On the right, if the previously suprathreshold excitatory input (E3) is given shortly after the inhibitory input (I), the IPSP prevents the excitatory potential from reaching threshold. The first systematic analysis of synaptic potentials in the CNS was in the early 1950s by Eccles and associates, who recorded intracellularly from spinal motor neurons. When a microelectrode enters a cell, there is a sudden change in the potential recorded by the electrode, which is typically about −60 mV (Figure 21-3). This is the resting membrane potential of the neuron. Two types of pathways— excitatory and inhibitory—impinge on the motor neuron. When an excitatory pathway is stimulated, a small depolarization or excitatory postsynaptic potential (EPSP) is recorded. This potential is due to the excitatory transmitter acting on an ionotropic receptor, causing an increase in cation permeability. As additional excitatory synapses are activated, there is a graded summation of the EPSPs to increase the size of the depolarization (Figure 21–3, top, spatial summation, middle). When a sufficient number of excitatory synapses are activated, the excitatory postsynaptic potential depolarizes the postsynaptic cell to threshold, and an all-or-none action potential is generated. Alternatively, if there is a repetitive firing of an excitatory input, the temporal summation of the EPSPs may also reach the action potential threshold (Figure 21–3, top, right). When an inhibitory pathway is stimulated, the postsynaptic membrane is hyperpolarized owing to the selective opening of chloride channels, producing an inhibitory postsynaptic potential (IPSP) (Figure 21–3, bottom, middle). However, because the equilibrium potential for chloride (see Chapter 14) is only slightly more negative than the resting potential (~ −65 mV), the hyperpolarization is small and contributes only modestly to the inhibitory action. The opening of the chloride channel during the inhibitory postsynaptic potential makes the neuron “leaky” so that changes in membrane potential are more difficult to achieve. This shunting effect decreases the change in membrane potential during the excitatory postsynaptic potential. As a result, an excitatory postsynaptic potential that evoked an action potential under resting conditions fails to evoke an action potential during the inhibitory postsynaptic potential (Figure 21–3, bottom, right). A second type of inhibition is presynaptic inhibition. It was first described for sensory fibers entering the spinal cord, where excitatory synaptic terminals receive synapses called axoaxonic synapses (described later). When activated, axoaxonic synapses reduce the amount of transmitter released from the terminals of sensory fibers. It is interesting that presynaptic inhibitory receptors are present


on almost all presynaptic terminals in the brain even though axoaxonic synapses appear to be restricted to the spinal cord. In the brain, transmitter can spill out of the synapse and activate presynaptic receptors, either on the same synapse (autoreceptors) or on neighboring synapses.

SITES OF DRUG ACTION Virtually all the drugs that act in the CNS produce their effects by modifying some step in chemical synaptic transmission. Figure 21–4 illustrates some of the steps that can be altered. These transmitter-dependent actions can be divided into presynaptic and postsynaptic categories.

FIGURE 21–4 Sites of drug action. Schematic drawing of steps at which drugs can alter synaptic transmission. (1) Action potential in presynaptic fiber; (2) synthesis of transmitter; (3) storage; (4) metabolism; (5) release; (6) reuptake into the nerve ending or uptake into a glial cell; (7) degradation; (8) receptor for the transmitter; (9) receptor-induced increase or decrease in ionic conductance; (10) retrograde signaling. Drugs acting on the synthesis, storage, metabolism, and release of neurotransmitters fall into the presynaptic category. Synaptic transmission can be depressed by blockade of transmitter synthesis or storage. For example, reserpine depletes monoamine synapses of transmitters by interfering with intracellular storage. Blockade of transmitter catabolism inside the nerve terminal can increase transmitter concentrations and has been reported to increase the amount of transmitter released per impulse. Drugs can also alter the release of


transmitters. The stimulant amphetamine induces the release of catecholamines from adrenergic synapses (see Chapters 6, 9, and 32). Capsaicin causes the release of the peptide substance P from sensory neurons, and tetanus toxin blocks the release of transmitters. After a CNS transmitter has been released into the synaptic cleft, its action is terminated either by uptake or by degradation. For most neurotransmitters, there are uptake mechanisms into the synaptic terminal and also into surrounding neuroglia. Cocaine, for example, blocks the uptake of catecholamines at adrenergic synapses and thus potentiates the action of these amines. Acetylcholine, however, is inactivated by enzymatic degradation, not reuptake. Anticholinesterases block the degradation of acetylcholine and thereby prolong its action (see Chapter 7). No uptake mechanism has been found for any of the numerous CNS peptides, and it has yet to be demonstrated whether specific enzymatic degradation terminates the action of peptide transmitters. In the postsynaptic region, the transmitter receptor provides the primary site of drug action. Drugs can act either as neurotransmitter agonists, such as the opioids, which mimic the action of enkephalin, or they can block receptor function. Receptor antagonism is a common mechanism of action for CNS drugs. An example is strychnine’s blockade of the receptor for the inhibitory transmitter glycine. This block, which underlies strychnine’s convulsant action, illustrates how the blockade of inhibitory processes results in excitation. Drugs can also act directly on the ion channel of ionotropic receptors. For example, the anesthetic ketamine blocks the NMDA subtype of glutamate ionotropic receptors by binding in the ion channel pore. In the case of metabotropic receptors, drugs can act at any of the steps downstream of the receptor. Perhaps the best example is provided by the methylxanthines, which can modify neurotransmitter responses mediated through the second-messenger cAMP. At high concentrations, the methylxanthines elevate the level of cAMP by blocking its metabolism and thereby prolong its action. The traditional view of the synapse is that it functions like a valve, transmitting information in one direction. However, it is now clear that the synapse can also generate signals that feed back onto the presynaptic terminal to modify transmitter release. Endocannabinoids are the best documented example of such retrograde signaling. Postsynaptic activity leads to the synthesis and release of endocannabinoids, which then bind to receptors on the presynaptic terminal. Although the gas nitric oxide (NO) has long been proposed as a retrograde messenger, its physiologic role in the CNS is still not well understood. The selectivity of CNS drug action is based on two primary factors. First, with a few exceptions, different neurotransmitters are released by different groups of neurons. These transmitters are often segregated into neuronal systems that subserve broadly different CNS functions. That this segregation occurs has provided neuroscientists with a powerful pharmacologic approach for analyzing CNS function and treating pathologic conditions. Second, there is a multiplicity of receptors for each neurotransmitter. For example, at least 14 different serotonin receptors are encoded by different genes. These receptors also often have differential cellular distributions throughout the CNS, allowing for the development of drugs that selectively target particular receptors and CNS functions.

CELLULAR ORGANIZATION OF THE BRAIN Most of the neuronal systems in the CNS can be divided into two broad categories: hierarchical systems and nonspecific or diffuse neuronal systems.

Hierarchical Systems Hierarchical systems include all the pathways directly involved in sensory perception and motor control. These pathways are generally clearly delineated, being composed of large myelinated fibers that can often conduct action potentials at a rate of more than 50 m/s. The information is typically phasic and occurs in bursts of action potentials. In sensory systems, the information is processed sequentially by successive integrations at each relay nucleus on its way to the cortex. A lesion at any link incapacitates the system. Within each nucleus and in the cortex, there are two types of cells: relay or projection neurons and local circuit neurons (Figure 21–5A). The projection neurons form the interconnecting pathways that transmit signals over long distances. Their cell bodies are relatively large, and their axons can project long distances but also emit small collaterals that synapse onto local interneurons. These neurons are excitatory, and their synaptic influences, which involve ionotropic receptors, are very short-lived. The excitatory transmitter released from these cells is, in most instances, glutamate.


FIGURE 21–5 Hierarchical pathways in the CNS. A shows parts of three excitatory relay neurons (blue) and two types of local inhibitory interneuron pathways, recurrent and feed-forward. The inhibitory neurons are shown in gray. B shows the pathway responsible for axoaxonic presynaptic inhibition in which the axon of an inhibitory neuron (gray) synapses onto the presynaptic axon terminal of an excitatory fiber (blue) to inhibit its neurotransmitter release. Local circuit neurons are typically smaller than projection neurons, and their axons arborize in the immediate vicinity of the cell body. Most of these neurons are inhibitory, and they release either GABA or glycine. They synapse primarily on the cell body of the projection neurons but can also synapse on the dendrites of projection neurons as well as with each other. Two common types of pathways for these neurons (Figure 21–5A) include recurrent feedback pathways and feed-forward pathways. A special class of local circuit neurons in the spinal cord forms axoaxonic synapses on the terminals of sensory axons (Figure 21–5B). Although there are a great variety of synaptic connections in these hierarchical systems, the fact that a limited number of transmitters are used by these neurons indicates that any major pharmacologic manipulation of this system will have a profound effect on the overall excitability of the CNS. For instance, selectively blocking GABAA receptors with a drug such as picrotoxin results in generalized convulsions. Thus,


although the mechanism of action of picrotoxin is specific in blocking the effects of GABA, the overall functional effect appears to be quite nonspecific, because GABA-mediated synaptic inhibition is so widely utilized in the brain.

Nonspecific or Diffuse Neuronal Systems Neuronal systems containing many of the other neurotransmitters, including the monoamines and acetylcholine, differ in fundamental ways from the hierarchical systems. These neurotransmitters are produced by only a limited number of neurons whose cell bodies are located in small discrete nuclei, often in the brainstem. For example, noradrenergic cell bodies are found primarily in a compact cell group, called the locus caeruleus, located in the caudal pontine central gray matter and number only approximately 1500 neurons on each side of the brain in the rat. However, from these limited nuclei, these neurons project widely and diffusely throughout the brain and spinal cord (Figure 21–6). Because the axons from these diffusely projecting neurons are fine and unmyelinated, they conduct very slowly, at about 0.5 m/s. The axons branch repeatedly and are extraordinarily divergent. Branches from the same neuron can innervate several functionally different parts of the CNS, synapsing onto and modulating neurons within the hierarchical systems. In the neocortex, these fibers have a tangential organization and therefore can influence large areas of cortex. In addition, most neurotransmitters utilized by diffuse neuronal systems, including norepinephrine, act predominantly on metabotropic receptors and therefore initiate long-lasting synaptic effects. Based on these observations, it is clear that the monoamine systems cannot be conveying topographically specific types of information; rather, vast areas of the CNS must be affected simultaneously and in a rather uniform way. It is not surprising, then, that these systems have been implicated in such global functions as sleeping and waking, attention, appetite, and emotional states.


FIGURE 21–6 Diffuse neurotransmitter pathways in the CNS. For each of the neurotransmitter pathways shown, the cell bodies are located in discrete brainstem or basal forebrain nuclei and project widely throughout the CNS. These diffuse systems largely modulate the function of the hierarchical pathways. Serotonin neurons, for example, are found in the midline raphe nuclei in the forebrain and send extraordinarily divergent projections to nearly all regions of the CNS. Other diffusely projecting neurotransmitter pathways include the histamine and orexin systems (not shown). VTA, ventral tegmental area; SN, substantia nigra; A1-A7, adrenergic brainstem nuclei; MSN, medial septal nucleus; DB, diagonal band of Broca; C5-C8, cholinergic brainstem nuclei.


CENTRAL NEUROTRANSMITTERS Because drug selectivity is based on the fact that different pathways use different transmitters, a primary goal of neuroscientists has been to identify the neurotransmitters in CNS pathways. Establishing that a chemical substance is a transmitter has been far more difficult for central synapses than for peripheral synapses. The following criteria have been established for transmitter identification. 1. Localization: A suspected transmitter must reside in the presynaptic terminal of the pathway of interest. 2. Release: A suspected transmitter must be released from a neuron in response to neuronal activity and in a calcium-dependent manner. 3. Synaptic mimicry: Application of the candidate substance should produce a response that mimics the action of the transmitter released by nerve stimulation, and application of a selective antagonist should block the response. Using the criteria above, a vast number of small molecules have been isolated from the brain, and studies using a variety of approaches suggest that the agents listed in Table 21–2 are neurotransmitters. A brief summary of these compounds follows. TABLE 21–2 Summary of neurotransmitter pharmacology in the central nervous system.



Amino Acid Neurotransmitters The amino acids of primary interest to the pharmacologist fall into two categories: the acidic amino acid glutamate and the neutral amino acids glycine and GABA. All three compounds are present in high concentrations in the CNS and are extremely potent modifiers of neuronal excitability. A. Glutamate Excitatory synaptic transmission is mediated by glutamate, which is present in very high concentrations in excitatory synaptic vesicles (~100 mM). Glutamate is released into the synaptic cleft by Ca2+-dependent exocytosis. The released glutamate acts on postsynaptic glutamate receptors and is cleared by glutamate transporters present on surrounding glia (Figure 21–7). In glia, glutamate is converted to glutamine by glutamine synthetase, released from the glia, taken up by the nerve terminal, and converted back to glutamate by the enzyme glutaminase. The high concentration of glutamate in synaptic vesicles is achieved by the vesicular glutamate transporter (VGLUT).


FIGURE 21–7 Schematic diagram of a glutamate synapse. Glutamine is imported into the glutamatergic neuron (A) and converted into glutamate by glutaminase. The glutamate is then concentrated in vesicles by the vesicular glutamate transporter. Upon release into the synapse, glutamate can interact with AMPA and NMDA ionotropic receptor channels (AMPAR, NMDAR) and with metabotropic receptors (mGluR) on the postsynaptic cell (B). Synaptic transmission is terminated by active transport of the glutamate into a neighboring glial cell (C) by a glutamate transporter. It is converted into glutamine by glutamine synthetase and transported back into the glutamatergic axon terminal. Almost all neurons that have been tested are strongly excited by glutamate. This excitation is caused by the activation of both ionotropic and metabotropic receptors, which have been extensively characterized by molecular cloning. The ionotropic receptors are divided into three subtypes based on the action of selective agonists: α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), kainic acid (KA), and N-methyl-D-aspartate (NMDA). All the ionotropic receptors are composed of four subunits. AMPA receptors, which are present on all neurons, are heterotetramers assembled from four subunits (GluA1–GluA4). The majority of AMPA receptors contain the GluA2 subunit and are permeable to Na+ and K+, but not to Ca2+. Some AMPA receptors, typically present on inhibitory interneurons, lack the GluA2 subunit and are also permeable to Ca2+. Kainate receptors are not as uniformly distributed as AMPA receptors, being expressed at high levels in the hippocampus, cerebellum, and spinal cord. They are formed from a number of subunit combinations (GluK1–GluK5). Although GluK4 and GluK5 are


unable to form channels on their own, their presence in the receptor changes the receptor’s affinity and kinetics. Similar to AMPA receptors, kainate receptors are permeable to Na+ and K+ and in some subunit combinations can also be permeable to Ca2+. NMDA receptors are as ubiquitous as AMPA receptors, being present on essentially all neurons in the CNS. All NMDA receptors require the presence of the subunit GluN1. The channel also contains one or two GluN2 subunits (GluN2A–GluN2D). Unlike AMPA and kainate receptors, all NMDA receptors are highly permeable to Ca2+ as well as to Na+ and K+. NMDA receptor function is controlled in a number of intriguing ways. In addition to glutamate binding, the channel also requires the binding of glycine to a separate site. The physiologic role of glycine binding is unclear because the glycine site appears to be saturated at normal ambient levels of glycine. Another important feature is that while AMPA and kainate receptor activation results in channel opening at resting membrane potential, NMDA receptor activation does not. This is due to the voltage-dependent block of the NMDA pore by extracellular Mg2+. Only when the neuron is strongly depolarized, as occurs with intense activation of the synapse or by activation of neighboring synapses, is Mg2+ expelled and the channel opened. Thus, there are two requirements for NMDA receptor channel opening: Glutamate must bind the receptor and the membrane must be depolarized. The rise in intracellular Ca2+ that accompanies channel opening results in a long-lasting enhancement in synaptic strength that is referred to as long-term potentiation (LTP). The change, which is one major type of synaptic plasticity, can last for many hours or even days and is generally accepted as an important cellular mechanism underlying learning and memory. The metabotropic glutamate receptors are G protein-coupled receptors that act indirectly on ion channels via G proteins. Metabotropic receptors (mGluR1–mGluR8) have been divided into three groups (I, II, and III). A variety of agonists and antagonists have been developed that interact selectively with the different groups. Group I receptors are typically located postsynaptically and activate phospholipase C, leading to inositol trisphosphate-mediated intracellular Ca2+ release. In contrast, group II and group III receptors are typically located on presynaptic nerve terminals and act as inhibitory autoreceptors. Activation of these receptors causes the inhibition of Ca2+ channels, resulting in inhibition of transmitter release. These receptors are activated only when the concentration of glutamate rises to high levels during repetitive stimulation of the synapse. Activation of these receptors also causes the inhibition of adenylyl cyclase and decreases cAMP generation. B. GABA and Glycine Both GABA and glycine are inhibitory neurotransmitters, which are typically released from local interneurons. Interneurons that release glycine are restricted to the spinal cord and brainstem, whereas interneurons releasing GABA are present throughout the CNS, including the spinal cord. It is interesting that some interneurons in the spinal cord can release both GABA and glycine. Glycine receptors are pentameric structures that are selectively permeable to Cl−. Strychnine, which is a potent spinal cord convulsant and has been used in some rat poisons, selectively blocks glycine receptors. GABA receptors are divided into two main types: GABA A and GABAB. Inhibitory postsynaptic potentials in many areas of the brain have a fast and slow component. The fast component is mediated by GABAA receptors and the slow component by GABAB receptors. The difference in kinetics stems from the differences in coupling of the receptors to ion channels. GABAA receptors are ionotropic receptors and, like glycine receptors, are pentameric structures that are selectively permeable to Cl−. These receptors are selectively inhibited by picrotoxin and bicuculline, both of which cause generalized convulsions. A great many subunits for GABA A receptors have been cloned; this accounts for the large diversity in the pharmacology of GABAA receptors, making them key targets for clinically useful agents (see Chapter 22). GABAB receptors are metabotropic receptors that are selectively activated by the antispastic drug baclofen. These receptors are coupled to G proteins that, depending on their cellular location, either inhibit Ca2+ channels or activate K+ channels. The GABAB component of the inhibitory postsynaptic potential is due to a selective increase in K+ conductance. This inhibitory postsynaptic potential is long-lasting and slow because the coupling of receptor activation to K+ channel opening is indirect and delayed. GABAB receptors are localized to the perisynaptic region and thus require the spillover of GABA from the synaptic cleft. GABA B receptors are also present on the axon terminals of many excitatory and inhibitory synapses. In this case, GABA spills over onto these presynaptic GABAB receptors, inhibiting transmitter release by inhibiting Ca2+ channels. In addition to their coupling to ion channels, GABAB receptors also inhibit adenylyl cyclase and decrease cAMP generation.

Acetylcholine Acetylcholine was the first compound to be identified pharmacologically as a transmitter in the CNS. Eccles showed in the early 1950s that excitation of spinal cord Renshaw cells by recurrent axon collaterals from spinal motor neurons was blocked by nicotinic antagonists. Furthermore, Renshaw cells were extremely sensitive to nicotinic agonists. This early success at identifying a transmitter for a central synapse was followed by disappointment because it remained the sole central synapse for which the transmitter was known until the late 1960s, when comparable data became available for GABA and glycine. The motor axon collateral synapse remains one of the bestdocumented examples of a cholinergic nicotinic synapse in the mammalian CNS, despite the rather widespread distribution of nicotinic receptors as defined by in situ hybridization studies.


Most CNS responses to acetylcholine are mediated by a large family of G protein-coupled muscarinic receptors. At a few sites, acetylcholine causes slow inhibition of the neuron by activating the M2 subtype of receptor, which opens potassium channels. A far more widespread muscarinic action in response to acetylcholine is a slow excitation that in some cases is mediated by M1 receptors. These muscarinic effects are much slower than either nicotinic effects on Renshaw cells or the effect of amino acids. Furthermore, this M1 muscarinic excitation is unusual in that acetylcholine produces it by decreasing the membrane permeability to potassium, ie, the opposite of conventional transmitter action. Eight major CNS nuclei of acetylcholine neurons have been characterized with diffuse projections. These include neurons in the neostriatum, the medial septal nucleus, and the reticular formation that appear to play an important role in cognitive functions, especially memory. Presenile dementia of the Alzheimer type is reportedly associated with a profound loss of cholinergic neurons. However, the specificity of this loss has been questioned because the levels of other putative transmitters, eg, somatostatin, are also decreased.

Monoamine Neurotransmitters Monoamines include the catecholamines (dopamine and norepinephrine) and 5-hydroxytryptamine. The diamine neurotransmitter, histamine, has several similarities to these monoamines. Although these compounds are present in very small amounts in the CNS, they can be localized using extremely sensitive histochemical methods. These pathways are the site of action of many drugs; for example, the CNS stimulants cocaine and amphetamine appear to act primarily at catecholamine synapses. Cocaine blocks the reuptake of dopamine and norepinephrine, whereas amphetamines cause presynaptic terminals to release these transmitters. A. Dopamine The major pathways containing dopamine are the projection linking the substantia nigra to the neostriatum and the projection linking the ventral tegmental region to limbic structures, particularly the limbic cortex. The therapeutic action of the antiparkinsonism drug levodopa is associated with the former area (see Chapter 28), whereas the therapeutic action of the antipsychotic drugs is thought to be associated with the latter (see Chapter 29). In addition, dopamine-containing neurons in the ventral hypothalamus play an important role in regulating pituitary function. Five dopamine receptors have been identified, and they fall into two categories: D1 -like (D1 and D5 ) and D2 -like (D2 , D3 , D4 ). All dopamine receptors are metabotropic. Dopamine generally exerts a slow inhibitory action on CNS neurons. This action has been best characterized on dopamine-containing substantia nigra neurons, where D2 -receptor activation opens potassium channels via the Gi coupling protein. B. Norepinephrine Most noradrenergic neurons are located in the locus caeruleus or the lateral tegmental area of the reticular formation. Although the density of fibers innervating various sites differs considerably, most regions of the CNS receive diffuse noradrenergic input. All noradrenergic receptor subtypes are metabotropic. When applied to neurons, norepinephrine can hyperpolarize them by increasing potassium conductance. This effect is mediated by ι2 receptors and has been characterized most thoroughly on locus caeruleus neurons. In many regions of the CNS, norepinephrine actually enhances excitatory inputs by both indirect and direct mechanisms. The indirect mechanism involves disinhibition; that is, inhibitory local circuit neurons are inhibited. The direct mechanism involves blockade of potassium conductances that slow neuronal discharge. Depending on the type of neuron, this effect is mediated by either ι1 or β receptors. Facilitation of excitatory synaptic transmission is in accordance with many of the behavioral processes thought to involve noradrenergic pathways, eg, attention and arousal. C. 5-Hydroxytryptamine Most 5-hydroxytryptamine (5-HT, serotonin) pathways originate from neurons in the midline raphe nuclei of the pons and upper brainstem. 5-HT is contained in unmyelinated fibers that diffusely innervate most regions of the CNS, but the density of the innervation varies. 5-HT acts on more than a dozen receptor subtypes. Except for the 5-HT3 receptor, all of these receptors are metabotropic. The ionotropic 5-HT3 receptor exerts a rapid excitatory action at a very limited number of sites in the CNS. In most areas of the CNS, 5-HT has a strong inhibitory action. This action is mediated by 5-HT1A receptors and is associated with membrane hyperpolarization caused by an increase in potassium conductance. It has been found that 5-HT1A receptors and GABAB receptors activate the same population of potassium channels. Some cell types are slowly excited by 5-HT owing to its blockade of potassium channels via 5-HT2 or 5-HT4 receptors. Both excitatory and inhibitory actions can occur on the same neuron. 5-HT has been implicated in the regulation of virtually all brain functions, including perception, mood, anxiety, pain, sleep, appetite, temperature, neuroendocrine control, and aggression. Given the broad roles of 5-HT in CNS function and the rich molecular diversity of 5-HT receptors, it is not surprising that many therapeutic agents target the 5-HT system (see Chapters 16, 29, 30, and 32). D. Histamine


In the CNS, histamine is exclusively made by neurons in the tuberomammillary nucleus (TMN) in the posterior hypothalamus. These neurons project widely throughout the brain and spinal cord where they modulate arousal, attention, feeding behavior, and memory (see Chapter 16). There are four histamine receptors (H1 to H4 ), all of which are metabotropic. Centrally acting antihistamines are generally used for their sedative properties and antagonism of H1 receptors is a common side effect of many drugs including some tricyclic antidepressants and antipsychotics.

Neuropeptides A great many CNS peptides have been discovered that produce dramatic effects both on animal behavior and on the activity of individual neurons. In many cases, peptide hormones discovered in the periphery (see Chapter 17) also act as neurotransmitters in the CNS. As most of these peptides were initially named according to their peripheral functions, the names are often unrelated to their CNS function. The pathways for many of the peptides have been mapped with immunohistochemical techniques and include opioid peptides (eg, enkephalins, endorphins), neurotensin, substance P, somatostatin, cholecystokinin, vasoactive intestinal polypeptide, neuropeptide Y, and thyrotropin-releasing hormone. Unlike the classical neurotransmitters above, which are packaged in small synaptic vesicles, neuropeptides are generally packaged in large, dense core vesicles. As in the peripheral autonomic nervous system, peptides often coexist with a conventional nonpeptide transmitter in the same neuron, but the release of the neuropeptides and the small molecule neurotransmitters can be independently regulated. Released neuropeptides may act locally or may diffuse long distances and bind to distant receptors. Most neuropeptide receptors are metabotropic and, like monoamine receptors, primarily serve modulatory roles in the nervous system. Neuropeptides have been implicated in a wide range of CNS functions including reproduction, social behaviors, appetite, arousal, pain, reward, and learning and memory. Thus, neuropeptides and their receptors are active targets of drug discovery efforts. A good example of the approaches used to define the role of these peptides in the CNS comes from studies on substance P and its association with sensory fibers. Substance P is contained in and released from small unmyelinated primary sensory neurons in the spinal cord and brainstem and causes a slow excitatory postsynaptic potential in target neurons. These sensory fibers are known to transmit noxious stimuli, and it is therefore surprising that—although substance P receptor antagonists can modify responses to certain types of pain—they do not block the response. Glutamate, which is released with substance P from these synapses, presumably plays an important role in transmitting pain stimuli. Substance P is certainly involved in many other functions because it is found in many areas of the CNS that are unrelated to pain pathways.

Orexin Orexins are peptide neurotransmitters produced in neurons in the lateral and posterior hypothalamus that, like the monoamine systems, project widely throughout the CNS. Orexins are also called hypocretins because of the near simultaneous discovery by two independent laboratories. Like most neuropeptides, orexin is released from large, dense core vesicles and bind to two G protein-coupled receptors, OX1 and OX2 . Orexin neurons also release glutamate and are thus excitatory. The orexin system, like the monoamine systems, projects widely throughout the CNS to influence physiology and behavior. In particular, orexin neurons exhibit firing patterns associated with wakefulness and project to and activate monoamine and acetylcholine neurons involved in sleep-wake cycles (see also Chapter 22). Animals lacking orexin or its receptors have narcolepsy and disrupted sleep-wake patterns. In addition to promoting wakefulness, the orexin system is involved in energy homeostasis, feeding behaviors, autonomic function, and reward.

Other Signaling Substances A. Endocannabinoids The primary psychoactive ingredient in cannabis, δ9 -tetrahy-drocannabinol (δ9 -THC), affects the brain mainly by activating a specific cannabinoid receptor, CB 1 . CB1 receptors are expressed at high levels in many brain regions, and they are primarily located on presynaptic terminals. Several endogenous brain lipids, including anandamide and 2-arachidonylglycerol (2-AG), have been identified as CB1 ligands. These ligands are not stored, as are classic neurotransmitters, but instead are rapidly synthesized by neurons in response to depolarization and consequent calcium influx. Activation of metabotropic receptors (eg, by acetylcholine and glutamate) can also activate the formation of 2-AG. In further contradistinction to classic neurotransmitters, endogenous cannabinoids can function as retrograde synaptic messengers: They are released from postsynaptic neurons and travel backward across synapses, activating CB1 receptors on presynaptic neurons and suppressing transmitter release. This suppression can be transient or long lasting, depending on the pattern of activity. Cannabinoids may affect memory, cognition, and pain perception by this mechanism. B. Nitric Oxide The CNS contains a substantial amount of nitric oxide synthase (NOS) within certain classes of neurons. This neuronal NOS is an enzyme activated by calcium-calmodulin, and activation of NMDA receptors, which increases intracellular calcium, results in the


generation of nitric oxide. Although a physiologic role for nitric oxide has been clearly established for vascular smooth muscle, its role in synaptic transmission and synaptic plasticity remains controversial. Nitric oxide diffuses freely across membranes and thus has been hypothesized to be a retrograde messenger, although this has not been demonstrated conclusively. Perhaps the strongest case for a role of nitric oxide in neuronal signaling in the CNS is for long-term depression of synaptic transmission in the cerebellum. C. Purines Receptors for purines, particularly adenosine, ATP, UTP, and UDP, are found throughout the body, including the CNS. High concentrations of ATP are found in and released from catecholinergic synaptic vesicles, and ATP may be converted to adenosine extracellularly by nucleotidases. Adenosine in the CNS acts on metabotropic A 1 receptors. Presynaptic A 1 receptors inhibit calcium channels and inhibit release of both amino acid and monoamine transmitters. ATP co-released with other neurotransmitters can bind to two classes of receptors. The P2X family of ATP receptors comprises nonselective ligand-gated cation channels, whereas the P2Y family is metabotropic. The physiological roles for co-released ATP remain elusive, but pharmacological studies suggest these receptors are involved in memory, wakefulness, and appetite, and may play roles in multiple neuropsychiatric disorders.

REFERENCES Basbaum AI et al: Cellular and molecular mechanisms of pain. Cell 2009;139:267. Berger M, Gray JA, Roth BL: T he expanded biology of serotonin. Annu Rev Med 2009;60:355. Castillo PE et al: Endocannabinoid signaling and synaptic function. Neuron 2012;76:70. Catterall WA: Voltage-gated calcium channels. Cold Spring Harb Perspect Biol 2011;3:a003947. Catterall WA: Voltage-gated sodium channels at 60: Structure, function and pathophysiology. J Physiol 2012;590:2577. Daneman R: T he blood-brain barrier in health and disease. Ann Neurol 2012;72:648. Gotter AL et al: International Union of Basic and Clinical Pharmacology. LXXXVI. Orexin receptor function, nomenclature and pharmacology. Pharmacol Rev 2012;64:389. Hille B: Ionic Channels of Excitable Membranes, 3rd ed. Sinauer, 2001. Jan LY, Jan YN: Voltage-gated potassium channels and the diversity of electrical signalling. J Physiol 2012;590:2591. Lewis RJ et al: Conus venom peptide pharmacology. Pharmacol Rev 2012;64:259. Khakh BS, North RA: Neuromodulation by extracellular AT P and P2X receptors in the CNS. Neuron 2012;76:51. Mody I, Pearce RA: Diversity of inhibitory neurotransmission through GABA(A) receptors. T rends Neurosci 2004;27:569. Nestler EJ, Hyman SE, Malenka RC: Molecular Neuropharmacology: A Foundation for Clinical Neurosceince, 2nd ed. McGraw-Hill, 2009. Nicoll RA, Roche KW: Long-term potentiation: peeling the onion. Neuropharmacology 2013;74:18. SĂźdhof T C, Rizo J: Synaptic vesicle exocytosis. Cold Spring Harb Perspect Biol 2011;3:a005637. T raynelis SF et al: Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010;62:405.


CHAPTER

22 Sedative-Hypnotic Drugs* Anthony J. Trevor, PhD

CASE STUDY At her annual physical examination, a 53-year-old middle school teacher complains that she has been having difficulty falling asleep, and after falling asleep, she awakens several times during the night. These episodes now occur almost nightly and are interfering with her ability to teach. She has tried various over-the-counter sleep remedies, but they were of little help and she experienced “hangover” effects on the day following their use. Her general health is good, she is not overweight, and she takes no prescription drugs. She drinks decaffeinated coffee but only one cup in the morning; however, she drinks as many as 6 cans per day of diet cola. She drinks a glass of wine with her evening meal but does not like stronger spirits. What other aspects of this patient’s history would you like to know? What therapeutic measures are appropriate for this patient? What drug, or drugs, (if any) would you prescribe?

Assignment of a drug to the sedative-hypnotic class indicates that it is able to cause sedation (with concomitant relief of anxiety) or to encourage sleep (hypnosis). Because there is considerable chemical variation within the group, this drug classification is based on clinical uses rather than on similarities in chemical structure. Anxiety states and sleep disorders are common problems, and sedative-hypnotics are widely prescribed drugs worldwide.

BASIC PHARMACOLOGY OF SEDATIVE-HYPNOTICS An effective sedative (anxiolytic) agent should reduce anxiety and exert a calming effect. The degree of central nervous system (CNS) depression caused by a sedative should be the minimum consistent with therapeutic efficacy. A hypnotic drug should produce drowsiness and encourage the onset and maintenance of a state of sleep. Hypnotic effects involve more pronounced depression of the CNS than sedation, and this can be achieved with many drugs in this class simply by increasing the dose. Graded dose-dependent depression of CNS function is a characteristic of most sedative-hypnotics. However, individual drugs differ in the relationship between the dose and the degree of CNS depression. Two examples of such dose-response relationships are shown in Figure 22–1. The linear slope for drug A is typical of many of the older sedative-hypnotics, including the barbiturates and alcohols. With such drugs, an increase in dose higher than that needed for hypnosis may lead to a state of general anesthesia. At still higher doses, these sedative-hypnotics may depress respiratory and vasomotor centers in the medulla, leading to coma and death. Deviations from a linear dose-response relationship, as shown for drug B, require proportionately greater dosage increments to achieve CNS depression more profound than hypnosis. This appears to be the case for benzodiazepines and for certain newer hypnotics that have a similar mechanism of action.


FIGURE 22–1 Dose-response curves for two hypothetical sedative-hypnotics.

CHEMICAL CLASSIFICATION The benzodiazepines are widely used sedative-hypnotics. All of the structures shown in Figure 22–2 are 1,4-benzodiazepines, and most contain a carboxamide group in the 7-membered heterocyclic ring structure. A substituent in the 7 position, such as a halogen or a nitro group, is required for sedative-hypnotic activity. The structures of triazolam and alprazolam include the addition of a triazole ring at the 1,2-position.


FIGURE 22–2 Chemical structures of benzodiazepines.


The chemical structures of some older and less commonly used sedative-hypnotics, including several barbiturates, are shown in Figure 22–3. Glutethimide and meprobamate are of distinctive chemical structure but are practically equivalent to barbiturates in their pharmacologic effects. They are rarely used. The sedative-hypnotic class also includes compounds of simpler chemical structure, including ethanol (see Chapter 23) and chloral hydrate.

FIGURE 22–3 Chemical structures of some barbiturates and other sedative-hypnotics. Several drugs with novel chemical structures have been introduced more recently for use in sleep disorders. Zolpidem, an imidazopyridine; zaleplon, a pyrazolopyrimidine; and eszopiclone, a cyclopyrrolone (Figure 22–4), although structurally unrelated to benzodiazepines, share a similar mechanism of action, as described below. Eszopiclone is the (S) enantiomer of zopiclone, a hypnotic drug that has been available outside the United States since 1989. Ramelteon and tasimelteon, melatonin receptor agonists, are newer hypnotic drugs (see Box: Ramelteon). Buspirone is a slow-onset anxiolytic agent whose actions are quite different from those of conventional sedative-hypnotics (see Box: Buspirone).


FIGURE 22–4 Chemical structures of newer hypnotics. Other classes of drugs that exert sedative effects include antipsychotics (see Chapter 29), and many antidepressant drugs (see Chapter 30). The latter are currently used widely in management of chronic anxiety disorders. Certain antihistaminic agents including hydroxyzine and promethazine (see Chapter 16) are also sedating. These agents commonly also exert marked effects on the peripheral autonomic nervous system. Other antihistaminic drugs with hypnotic effects, eg, diphenhydramine and doxylamine, are available in overthe-counter sleep aids.

Pharmacokinetics A. Absorption and Distribution The rates of oral absorption of sedative-hypnotics differ depending on a number of factors, including lipophilicity. For example, the absorption of triazolam is extremely rapid, and that of diazepam and the active metabolite of clorazepate is more rapid than other commonly used benzodiazepines. Clorazepate, a prodrug, is converted to its active form, desmethyldiazepam (nordiazepam), by acid hydrolysis in the stomach. Most of the barbiturates and other older sedative-hypnotics, as well as the newer hypnotics (eszopiclone,


zaleplon, zolpidem), are absorbed rapidly into the blood following oral administration. Lipid solubility plays a major role in determining the rate at which a particular sedative-hypnotic enters the CNS. This property is responsible for the rapid onset of the effects of triazolam, thiopental (see Chapter 25), and the newer hypnotics. All sedative-hypnotics cross the placental barrier during pregnancy. If sedative-hypnotics are given during the predelivery period, they may contribute to the depression of neonatal vital functions. Sedative-hypnotics are also detectable in breast milk and may exert depressant effects in the nursing infant. B. Biotransformation Metabolic transformation to more water-soluble metabolites is necessary for clearance of sedative-hypnotics from the body. The microsomal drug-metabolizing enzyme systems of the liver are most important in this regard, so elimination half-life of these drugs depends mainly on the rate of their metabolic transformation.

Ramelteon and Tasimelteon Melatonin receptors are thought to be involved in maintaining circadian rhythms underlying the sleep-wake cycle (see Chapter 16). Ramelteon, a novel hypnotic drug prescribed specifically for patients who have difficulty in falling asleep, is an agonist at MT1 and MT2 melatonin receptors located in the suprachiasmatic nuclei of the brain. Tasimelteon is similar and is approved for non-24 hour sleep-wake disorder. These drugs have no direct effects on GABAergic neurotransmission in the central nervous system. In polysomnography studies of patients with chronic insomnia, ramelteon reduced the latency of persistent sleep with no effects on sleep architecture and no rebound insomnia or significant withdrawal symptoms. The drug is rapidly absorbed after oral administration and undergoes extensive first-pass metabolism, forming an active metabolite with longer half-life (2–5 hours) than the parent drug. The CYP1A2 isoform of cytochrome P450 is mainly responsible for the metabolism of ramelteon, but the CYP2C9 isoform is also involved. Ramelteon should not be used in combination with inhibitors of CYP1A2 (eg, ciprofloxacin, fluvoxamine, tacrine, zileuton) or CYP2C9 (eg, fluconazole). Concurrent use with the antidepressant fluvoxamine increases the peak plasma concentration of ramelteon over 50-fold! Ramelteon should be used with caution in patients with liver dysfunction. The CYP inducer rifampin markedly reduces the plasma levels of both ramelteon and its active metabolite. Adverse effects of ramelteon include dizziness, somnolence, fatigue, and endocrine changes. 1 . Benzodiazepines—Hepatic metabolism accounts for the clearance of all benzodiazepines. The patterns and rates of metabolism depend on the individual drugs. Most benzodiazepines undergo microsomal oxidation (phase I reactions), including N-dealkylation and aliphatic hydroxylation catalyzed by cytochrome P450 isozymes, especially CYP3A4. The metabolites are subsequently conjugated (phase II reactions) to form glucuronides that are excreted in the urine. However, many phase I metabolites of benzodiazepines are pharmacologically active, some with long half-lives (Figure 22–5). For example, desmethyldiazepam, which has an elimination half-life of more than 40 hours, is an active metabolite of chlordiazepoxide, diazepam, prazepam, and clorazepate. Alprazolam and triazolam undergo α-hydroxylation, and the resulting metabolites appear to exert short-lived pharmacologic effects because they are rapidly conjugated to form inactive glucuronides. The short elimination half-life of triazolam (2–3 hours) favors its use as a hypnotic rather than as a sedative drug.


FIGURE 22–5 Biotransformation of benzodiazepines. Boldface, drugs available for clinical use in various countries; *, active metabolite. The formation of active metabolites has complicated studies on the pharmacokinetics of the benzodiazepines in humans because the elimination half-life of the parent drug may have little relation to the time course of pharmacologic effects. Benzodiazepines for which the parent drug or active metabolites have long half-lives are more likely to cause cumulative effects with multiple doses. Cumulative and residual effects such as excessive drowsiness appear to be less of a problem with such drugs as estazolam, oxazepam, and lorazepam, which have relatively short half-lives and are metabolized directly to inactive glucuronides. Some pharmacokinetic properties of selected benzodiazepines and newer hypnotics are listed in Table 22–1. The metabolism of several commonly used benzodiazepines including diazepam, midazolam, and triazolam is affected by inhibitors and inducers of hepatic P450 isozymes (see Chapter 4). TABLE 22–1 Pharmacokinetic properties of some benzodiazepines and newer hypnotics in humans.


Buspirone Buspirone has selective anxiolytic effects, and its pharmacologic characteristics are different from those of other drugs described in this chapter. Buspirone relieves anxiety without causing marked sedative, hypnotic, or euphoric effects. Unlike benzodiazepines, the drug has no anticonvulsant or muscle relaxant properties. Buspirone does not interact directly with GABAergic systems. It may exert its anxiolytic effects by acting as a partial agonist at brain 5-HT1A receptors, but it also has affinity for brain dopamine D2 receptors. Buspirone-treated patients show no rebound anxiety or withdrawal signs on abrupt discontinuance. The drug is not effective in blocking the acute withdrawal syndrome resulting from abrupt cessation of use of benzodiazepines or other sedativehypnotics. Buspirone has minimal abuse liability. In marked contrast to the benzodiazepines, the anxiolytic effects of buspirone may take 3–4 weeks to become established, making the drug unsuitable for management of acute anxiety states. The drug is used in generalized anxiety states but is less effective in panic disorders. Buspirone is rapidly absorbed orally but undergoes extensive first-pass metabolism via hydroxylation and dealkylation reactions to form several active metabolites. The major metabolite is 1-(2-pyrimidyl)-piperazine (1-PP), which has ι2 -adrenoceptor-blocking actions and which enters the central nervous system to reach higher levels than the parent drug. It is not known what role (if any) 1-PP plays in the central actions of buspirone. The elimination half-life of buspirone is 2–4 hours, and liver dysfunction may slow its clearance. Rifampin, an inducer of cytochrome P450, decreases the half-life of buspirone; inhibitors of CYP3A4 (eg, erythromycin, ketoconazole, grapefruit juice, nefazodone) can markedly increase its plasma levels. Buspirone causes less psychomotor impairment than benzodiazepines and does not affect driving skills. The drug does not potentiate effects of conventional sedative-hypnotic drugs, ethanol, or tricyclic antidepressants; and elderly patients do not appear to


be more sensitive to its actions. Nonspecific chest pain, tachycardia, palpitations, dizziness, nervousness, headache, tinnitus, gastrointestinal distress, and paresthesias and a dose-dependent pupillary constriction may occur. Blood pressure may be significantly elevated in patients receiving MAO inhibitors. Buspirone is an FDA category B drug in terms of its use in pregnancy. 2. Barbiturates—With the exception of phenobarbital, only insignificant quantities of the barbiturates are excreted unchanged. The major metabolic pathways involve oxidation by hepatic enzymes to form alcohols, acids, and ketones, which appear in the urine as glucuronide conjugates. The overall rate of hepatic metabolism in humans depends on the individual drug but (with the exception of the thiobarbiturates) is usually slow. The elimination half-lives of secobarbital and pentobarbital range from 18 to 48 hours in different individuals. The elimination half-life of phenobarbital in humans is 4–5 days. Multiple dosing with these agents can lead to cumulative effects. 3. Newer hypnotics—After oral administration of the standard formulation, zolpidem reaches peak plasma levels in 1–3 hours (Table 221). Sublingual and oral spray formulations of zolpidem are also available. Zolpidem is rapidly metabolized to inactive metabolites via oxidation and hydroxylation by hepatic CYP3A4. The elimination half-life of the drug is greater in women and is increased significantly in the elderly. A biphasic extended-release formulation extends plasma levels by approximately 2 hours. Zaleplon is metabolized to inactive metabolites mainly by hepatic aldehyde oxidase and partly by the cytochrome P450 iso-form CYP3A4. Dosage should be reduced in patients with hepatic impairment and in the elderly. Cimetidine, which inhibits both aldehyde dehydrogenase and CYP3A4, markedly increases the peak plasma level of zaleplon. Eszopiclone is metabolized by hepatic cytochromes P450 (especially CYP3A4) to form the inactive N-oxide derivative and weakly active desmethyleszopiclone. The elimination half-life of eszopiclone is prolonged in the elderly and in the presence of inhibitors of CYP3A4 (eg, ketoconazole). Inducers of CYP3A4 (eg, rifampin) increase the hepatic metabolism of eszopiclone. C. Excretion The water-soluble metabolites of sedative-hypnotics, mostly formed via the phase II conjugation of phase I metabolites, are excreted mainly via the kidney. In most cases, changes in renal function do not have a marked effect on the elimination of parent drugs. Phenobarbital is excreted unchanged in the urine to a certain extent (20–30% in humans), and its elimination rate can be increased significantly by alkalinization of the urine. This is partly due to increased ionization at alkaline pH, since phenobarbital is a weak acid with a pKa of 7.4. D. Factors Affecting Biodisposition The biodisposition of sedative-hypnotics can be influenced by several factors, particularly alterations in hepatic function resulting from disease or drug-induced increases or decreases in microsomal enzyme activities (see Chapter 4). In very old patients and in patients with severe liver disease, the elimination half-lives of these drugs are often increased significantly. In such cases, multiple normal doses of these sedative-hypnotics can result in excessive CNS effects. The activity of hepatic microsomal drug-metabolizing enzymes may be increased in patients exposed to certain older sedativehypnotics on a long-term basis (enzyme induction; see Chapter 4). Barbiturates (especially phenobarbital) and meprobamate are most likely to cause this effect, which may result in an increase in their hepatic metabolism as well as that of other drugs. Increased biotransformation of other pharmacologic agents as a result of enzyme induction by barbiturates is a potential mechanism underlying drug interactions (see Chapter 66). In contrast, benzodiazepines and the newer hypnotics do not change hepatic drug-metabolizing enzyme activity with continuous use.

Pharmacodynamics of Benzodiazepines, Barbiturates, & Newer Hypnotics A. Molecular Pharmacology of the GABAAReceptor The benzodiazepines, the barbiturates, zolpidem, zaleplon, eszopiclone, and many other drugs bind to molecular components of the GABAA receptor in neuronal membranes in the CNS. This receptor, which functions as a chloride ion channel, is activated by the inhibitory neurotransmitter GABA (see Chapter 21). The GABAA receptor has a pentameric structure assembled from five subunits (each with four membrane-spanning domains) selected from multiple polypeptide classes (α, β, γ, δ, ε, π, ρ, etc). Multiple subunits of several of these classes have been characterized, eg, six different α, four β, and three γ. A model of the GABA A receptor-chloride ion channel macromolecular complex is shown in Figure 22–6.


FIGURE 22–6 A model of the GABAA receptor-chloride ion channel macromolecular complex. A hetero-oligomeric glycoprotein, the complex consists of five or more membrane-spanning subunits. Multiple forms of α, β, and γ subunits are arranged in different pentameric combinations so that GABAA receptors exhibit molecular heterogeneity. GABA appears to interact at two sites between α and β subunits triggering chloride channel opening with resulting membrane hyperpolarization. Binding of benzodiazepines and the newer hypnotic drugs such as zolpidem occurs at a single site between α and γ subunits, facilitating the process of chloride ion channel opening. The benzodiazepine antagonist flumazenil also binds at this site and can reverse the hypnotic effects of zolpidem. Note that these binding sites are distinct from those of the barbiturates. (See also text and Box: The Versatility of the Chloride Channel GABA Receptor Complex.) A major isoform of the GABA A receptor that is found in many regions of the brain consists of two α1 subunits, two β2 subunits, and one γ2 subunit. In this isoform, the two binding sites for GABA are located between adjacent α1 and β2 subunits, and the binding pocket for benzodiazepines (the BZ site of the GABAA receptor) is between an α1 and the γ2 subunit. However, GABA A receptors in different areas of the CNS consist of various combinations of the essential subunits, and the benzodiazepines bind to many of these, including receptor isoforms containing α2, α3, and α5 subunits. Barbiturates also bind to multiple isoforms of the GABAA receptor but at different sites from those with which benzodiazepines interact. In contrast to benzodiazepines, zolpidem, zaleplon, and eszopiclone bind more selectively because these drugs interact only with GABAA-receptor isoforms that contain α1 subunits. The heterogeneity of GABAA receptors may constitute the molecular basis for the varied pharmacologic actions of benzodiazepines and related drugs (see Box: GABA Receptor Heterogeneity & Pharmacologic Selectivity). In contrast to GABA itself, benzodiazepines and other sedative-hypnotics have a low affinity for GABA B receptors, which are activated by the spasmolytic drug baclofen (see Chapters 21 and 27). B. Neuropharmacology GABA (γ-aminobutyric acid) is a major inhibitory neurotransmitter in the CNS (see Chapter 21). Electrophysiologic studies have shown that benzodiazepines potentiate GABAergic inhibition at all levels of the neuraxis, including the spinal cord, hypothalamus, hippocampus, substantia nigra, cerebellar cortex, and cerebral cortex. Benzodiazepines appear to increase the efficiency of GABAergic synaptic inhibition. The benzodiazepines do not substitute for GABA but appear to enhance GABA’s effects allosterically without directly activating GABAA receptors or opening the associated chloride channels. The enhancement in chloride ion conductance induced by the interaction of benzodiazepines with GABA takes the form of an increase in the frequency of channel-opening events.


GABA Receptor Heterogeneity & Pharmacologic Selectivity Studies involving genetically engineered (“knockout”) rodents have demonstrated that the specific pharmacologic actions elicited by benzodiazepines and other drugs that modulate GABA actions are influenced by the composition of the subunits assembled to form the GABAA receptor. Benzodiazepines interact primarily with brain GABA A receptors in which the α subunits (1, 2, 3, and 5) have a conserved histidine residue in the N-terminal domain. Strains of mice, in which a point mutation has been inserted converting histidine to arginine in the α1 subunit, show resistance to both the sedative and amnestic effects of benzodiazepines, but anxiolytic and muscle-relaxing effects are largely unchanged. These animals are also unresponsive to the hypnotic actions of zolpidem and zaleplon, drugs that bind selectively to GABAA receptors containing α1 subunits. In contrast, mice with selective histidine-arginine mutations in the α2 or α3 subunits of GABAA receptors show selective resistance to the antianxiety effects of benzodiazepines. Based on studies of this type, it has been suggested that α1 subunits in GABAA receptors mediate sedation, amnesia, and ataxic effects of benzodiazepines, whereas α2 and α3 subunits are involved in their anxiolytic and muscle-relaxing actions. Other mutation studies have led to suggestions that an α5 subtype is involved in at least some of the memory impairment caused by benzodiazepines. It should be emphasized that these studies involving genetic manipulations of the GABAA receptor utilize rodent models of the anxiolytic and amnestic actions of drugs. Barbiturates also facilitate the actions of GABA at multiple sites in the CNS, but—in contrast to benzodiazepines—they appear to increase the duration of the GABA-gated chloride channel openings. At high concentrations, the barbiturates may also be GABAmimetic, directly activating chloride channels. These effects involve a binding site or sites distinct from the benzodiazepine binding sites. Barbiturates are less selective in their actions than benzodiazepines, because they also depress the actions of the excitatory neurotransmitter glutamic acid via binding to the AMPA receptor. Barbiturates also exert nonsynaptic membrane effects in parallel with their effects on GABA and glutamate neurotransmission. This multiplicity of sites of action of barbiturates may be the basis for their ability to induce full surgical anesthesia (see Chapter 25) and for their more pronounced central depressant effects (which result in their low margin of safety) compared with benzodiazepines and the newer hypnotics. C. Benzodiazepine Binding Site Ligands The components of the GABAA receptor-chloride ion channel macromolecule that function as benzodiazepine binding sites exhibit heterogeneity (see Box: The Versatility of the Chloride Channel GABA Receptor Complex). Three types of ligand-benzodiazepine receptor interactions have been reported: (1) Agonists facilitate GABA actions, and this occurs at multiple BZ binding sites in the case of the benzodiazepines. As noted above, the nonbenzodiazepines zolpidem, zaleplon, and eszopiclone are selective agonists at the BZ sites that contain an α1 subunit. Endogenous agonist ligands for the BZ binding sites have been proposed, because benzodiazepine-like chemicals have been isolated from brain tissue of animals never exposed to these drugs. Nonbenzodiazepine molecules that have affinity for BZ sites on the GABAA receptor have also been detected in human brain. (2) Antagonists are typified by the synthetic benzodiazepine derivative flumazenil, which blocks the actions of benzodiazepines, eszopiclone, zaleplon, and zolpidem, but does not antagonize the actions of barbiturates, meprobamate, or ethanol. Certain endogenous neuropeptides are also capable of blocking the interaction of benzodiazepines with BZ binding sites. (3) Inverse agonists act as negative allosteric modulators of GABA-receptor function (see Chapter 1). Their interaction with BZ sites on the GABAA receptor can produce anxiety and seizures, an action that has been demonstrated for several compounds, especially the β-carbolines, eg, n-butyl-β-carboline-3-carboxylate (β-CCB). In addition to their direct actions, these molecules can block the effects of benzodiazepines. The physiologic significance of endogenous modulators of the functions of GABA in the CNS remains unclear. To date, it has not been established that the putative endogenous ligands of BZ binding sites play a role in the control of states of anxiety, sleep patterns, or any other characteristic behavioral expression of CNS function.

The Versatility of the Chloride Channel GABA Receptor Complex The GABAA-chloride channel macromolecular complex is one of the most versatile drug-responsive machines in the body. In addition to the benzodiazepines, barbiturates, and the newer hypnotics (eg, zolpidem), many other drugs with central nervous system effects can modify the function of this important ionotropic receptor. These include alcohol and certain intravenous anesthetics (etomidate, propofol) in addition to thiopental. For example, etomidate and propofol (see Chapter 25) appear to act selectively at GABAA receptors that contain a2 and a3 subunits, the latter suggested to be the most important with respect to the hypnotic and muscle-relaxing actions of these anesthetic agents. The anesthetic steroid alphaxalone is thought to interact with GABAA receptors, and they may also be targets for some of the actions of volatile anesthetics (eg, halothane). Most of these agents facilitate or mimic the action of GABA. However, it has not been shown that all these drugs act exclusively by this mechanism. Other drugs used in


the management of seizure disorders indirectly influence the activity of the GABAA-chloride channel macro-molecular complex by inhibiting GABA metabolism (eg, vigabatrin) or the reuptake of the transmitter (eg, tiagabine). Central nervous system excitatory agents that act on the chloride channel include picrotoxin and bicuculline. These convulsant drugs block the channel directly (picrotoxin) or interfere with GABA binding (bicuculline). D. Organ Level Effects 1. Sedation—Benzodiazepines, barbiturates, and most older sedative-hypnotic drugs exert calming effects with concomitant reduction of anxiety at relatively low doses. In most cases, however, the anxiolytic actions of sedative-hypnotics are accompanied by some depressant effects on psychomotor and cognitive functions. In experimental animal models, benzodiazepines and older sedative-hypnotic drugs are able to disinhibit punishment-suppressed behavior. This disinhibition has been equated with antianxiety effects of sedativehypnotics, and it is not a characteristic of all drugs that have sedative effects, eg, the tricyclic antidepressants and antihistamines. However, the disinhibition of previously suppressed behavior may be more related to behavioral disinhibitory effects of sedativehypnotics, including euphoria, impaired judgment, and loss of self-control, which can occur at dosages in the range of those used for management of anxiety. The benzodiazepines also exert dose-dependent anterograde amnesic effects (inability to remember events occurring during the drug’s duration of action). 2. Hypnosis—By definition, all of the sedative-hypnotics induce sleep if high enough doses are given. The effects of sedative-hypnotics on the stages of sleep depend on several factors, including the specific drug, the dose, and the frequency of its administration. The general effects of benzodiazepines and older sedative-hypnotics on patterns of normal sleep are as follows: (1) the latency of sleep onset is decreased (time to fall asleep); (2) the duration of stage 2 NREM (non-rapid eye movement) sleep is increased; (3) the duration of REM (rapid eye movement) sleep is decreased; and (4) the duration of stage 4 NREM slow-wave sleep is decreased. The newer hypnotics all decrease the latency to persistent sleep. Zolpidem decreases REM sleep but has minimal effect on slow-wave sleep. Zaleplon decreases the latency of sleep onset with little effect on total sleep time, NREM, or REM sleep. Eszopiclone increases total sleep time, mainly via increases in stage 2 NREM sleep, and at low doses has little effect on sleep patterns. At the highest recommended dose, eszopiclone decreases REM sleep. More rapid onset of sleep and prolongation of stage 2 are presumably clinically useful effects. However, the significance of older sedative-hypnotic drug effects on REM and slow-wave sleep is not clear. Deliberate interruption of REM sleep causes anxiety and irritability followed by a rebound increase in REM sleep at the end of the experiment. A similar pattern of “REM rebound” can be detected following abrupt cessation of drug treatment with older sedative-hypnotics, especially when drugs with short durations of action (eg, triazolam) are used at high doses. With respect to zolpidem and the other newer hypnotics, there is little evidence of REM rebound when these drugs are discontinued after use of recommended doses. However, rebound insomnia occurs with both zolpidem and zaleplon if used at higher doses. Despite possible reductions in slow-wave sleep, there are no reports of disturbances in the secretion of pituitary or adrenal hormones when either barbiturates or benzodiazepines are used as hypnotics. The use of sedative-hypnotics for more than 1–2 weeks leads to some tolerance to their effects on sleep patterns. 3. Anesthesia—As shown in Figure 22–1, high doses of certain sedative-hypnotics depress the CNS to the point known as stage III of general anesthesia (see Chapter 25). However, the suitability of a particular agent as an adjunct in anesthesia depends mainly on the physicochemical properties that determine its rapidity of onset and duration of effect. Among the barbiturates, thiopental and methohexital are very lipid-soluble, penetrating brain tissue rapidly following intravenous administration, a characteristic favoring their use for the induction of anesthesia. Rapid tissue redistribution (not rapid elimination) accounts for the short duration of action of these drugs, a feature useful in recovery from anesthesia. Benzodiazepines—including diazepam, lorazepam, and midazolam—are used intravenously in anesthesia (see Chapter 25), often in combination with other agents. Not surprisingly, benzodiazepines given in large doses as adjuncts to general anesthetics may contribute to a persistent postanesthetic respiratory depression. This is probably related to their relatively long half-lives and the formation of active metabolites. However, such depressant actions of the benzodiazepines are usually reversible with flumazenil. 4. Anticonvulsant effects—Most sedative-hypnotics are capable of inhibiting the development and spread of epileptiform electrical activity in the CNS. Some selectivity exists in that some members of the group can exert anticonvulsant effects without marked CNS depression (although psychomotor function may be impaired). Several benzodiazepines—including clonazepam, nitrazepam, lorazepam, and diazepam—are sufficiently selective to be clinically useful in the management of seizures (see Chapter 24). Of the barbiturates, phenobarbital and metharbital (converted to phenobarbital in the body) are effective in the treatment of generalized tonic-clonic seizures, though not the drugs of first choice. However, zolpidem, zaleplon, and eszopiclone lack anticonvulsant activity, presumably because of their more selective binding than that of benzodiazepines to GABAA receptor isoforms. 5. Muscle relaxation—Certain drugs in the sedative-hypnotic class, particularly members of the carbamate (eg, meprobamate) and benzodiazepine groups, exert inhibitory effects on polysynaptic reflexes and internuncial transmission and at high doses may also depress


transmission at the skeletal neuromuscular junction. Somewhat selective actions of this type that lead to muscle relaxation can be readily demonstrated in animals and have led to claims of usefulness for relaxing contracted voluntary muscle in muscle spasm (see Clinical Pharmacology). Muscle relaxation is not a characteristic action of zolpidem, zaleplon, and eszopiclone. 6. Effects on respiration and cardiovascular function—At hypnotic doses in healthy patients, the effects of sedative-hypnotics on respiration are comparable to changes during natural sleep. However, even at therapeutic doses, sedative-hypnotics can produce significant respiratory depression in patients with pulmonary disease. Effects on respiration are dose-related, and depression of the medullary respiratory center is the usual cause of death due to overdose of sedative-hypnotics. At doses up to those causing hypnosis, no significant effects on the cardiovascular system are observed in healthy patients. However, in hypovolemic states, heart failure, and other diseases that impair cardiovascular function, normal doses of sedative-hypnotics may cause cardiovascular depression, probably as a result of actions on the medullary vasomotor centers. At toxic doses, myocardial contractility and vascular tone may both be depressed by central and peripheral effects, possibly via facilitation of the actions of adenosine, leading to circulatory collapse. Respiratory and cardiovascular effects are more marked when sedative-hypnotics are given intravenously.

Tolerance: Psychologic & Physiologic Dependence Tolerance—decreased responsiveness to a drug following repeated exposure—is a common feature of sedative-hypnotic use. It may result in the need for an increase in the dose required to maintain symptomatic improvement or to promote sleep. It is important to recognize that partial cross-tolerance occurs between the sedative-hypnotics described here and also with ethanol (see Chapter 23)—a feature of some clinical importance, as explained below. The mechanisms responsible for tolerance to sedative-hypnotics are not well understood. An increase in the rate of drug metabolism (metabolic tolerance) may be partly responsible in the case of chronic administration of barbiturates, but changes in responsiveness of the CNS (pharmacodynamic tolerance) are of greater importance for most sedative-hypnotics. In the case of benzodiazepines, the development of tolerance in animals has been associated with downregulation of brain benzodiazepine receptors. Tolerance has been reported to occur with the extended use of zolpidem. Minimal tolerance was observed with the use of zaleplon over a 5-week period and eszopiclone over a 6-month period. The perceived desirable properties of relief of anxiety, euphoria, disinhibition, and promotion of sleep have led to the compulsive misuse of virtually all sedative-hypnotics. (See Chapter 32 for a detailed discussion.) For this reason, most sedative-hypnotic drugs are classified as Schedule III or Schedule IV drugs for prescribing purposes. The consequences of abuse of these agents can be defined in both psychologic and physiologic terms. The psychologic component may initially parallel simple neurotic behavior patterns difficult to differentiate from those of the inveterate coffee drinker or cigarette smoker. When the pattern of sedative-hypnotic use becomes compulsive, more serious complications develop, including physiologic dependence and tolerance. Physiologic dependence can be described as an altered physiologic state that requires continuous drug administration to prevent an abstinence or withdrawal syndrome. In the case of sedative-hypnotics, this syndrome is characterized by states of increased anxiety, insomnia, and CNS excitability that may progress to convulsions. Most sedative-hypnotics—including benzodiazepines—are capable of causing physiologic dependence when used on a long-term basis. However, the severity of withdrawal symptoms differs among individual drugs and depends also on the magnitude of the dose used immediately before cessation of use. When higher doses of sedative-hypnotics are used, abrupt withdrawal leads to more serious withdrawal signs. Differences in the severity of withdrawal symptoms resulting from individual sedative-hypnotics relate in part to half-life, since drugs with long half-lives are eliminated slowly enough to accomplish gradual withdrawal with few physical symptoms. The use of drugs with very short half-lives for hypnotic effects may lead to signs of withdrawal even between doses. For example, triazolam, a benzodiazepine with a half-life of about 4 hours, has been reported to cause daytime anxiety when used to treat sleep disorders. The abrupt cessation of zolpidem, zaleplon, or eszopiclone may also result in withdrawal symptoms, though usually of less intensity than those seen with benzodiazepines.

BENZODIAZEPINE ANTAGONISTS: FLUMAZENIL Flumazenil is one of several 1,4-benzodiazepine derivatives with a high affinity for the benzodiazepine binding site on the GABAA receptor that act as competitive antagonists. It blocks many of the actions of benzodiazepines, zolpidem, zaleplon, and eszopiclone, but does not antagonize the CNS effects of other sedative-hypnotics, ethanol, opioids, or general anesthetics. Flumazenil is approved for use in reversing the CNS depressant effects of benzodiazepine overdose and to hasten recovery following use of these drugs in anesthetic and diagnostic procedures. Although the drug reverses the sedative effects of benzodiazepines, antagonism of benzodiazepine-induced respiratory depression is less predictable. When given intravenously, flumazenil acts rapidly but has a short half-life (0.7–1.3 hours) due to rapid hepatic clearance. Because all benzodiazepines have a longer duration of action than flumazenil, sedation commonly recurs, requiring repeated administration of the antagonist.

Orexin Receptor Antagonists: Sleep-Enabling Drugs


Orexin A and B are peptides found in hypothalamic neurons that are involved in the control of wakefulness; their levels increase in the day and decrease at night. Loss of orexin neurons is associated with narcolepsy, a disorder characterized by daytime sleepiness and cataplexy. Animal studies show that orexin receptor antagonists have sleep-enabling effects. This has prompted the development of a new class of hypnotic drugs, orexin antagonists, which include the drugs almorexant and suvorexant, the latter agent recently approved by the FDA. Adverse effects of flumazenil include agitation, confusion, dizziness, and nausea. Flumazenil may cause a severe precipitated abstinence syndrome in patients who have developed physiologic benzodiazepine dependence. In patients who have ingested benzodiazepines with tricyclic antidepressants, seizures and cardiac arrhythmias may follow flumazenil administration.

CLINICAL PHARMACOLOGY OF SEDATIVE-HYPNOTICS TREATMENT OF ANXIETY STATES The psychologic, behavioral, and physiologic responses that characterize anxiety can take many forms. Typically, the psychic awareness of anxiety is accompanied by enhanced vigilance, motor tension, and autonomic hyperactivity. Anxiety is often secondary to organic disease states—acute myocardial infarction, angina pectoris, gastrointestinal ulcers, etc—which themselves require specific therapy. Another class of secondary anxiety states (situational anxiety) results from circumstances that may have to be dealt with only once or a few times, including anticipation of frightening medical or dental procedures and family illness or other stressful event. Even though situational anxiety tends to be self-limiting, the short-term use of sedative-hypnotics may be appropriate for the treatment of this and certain disease-associated anxiety states. Similarly, the use of a sedative-hypnotic as premedication prior to surgery or some unpleasant medical procedure is rational and proper (Table 22–2). TABLE 22–2 Clinical uses of sedative-hypnotics.

Excessive or unreasonable anxiety about life circumstances (generalized anxiety disorder, GAD), panic disorders, and agoraphobia are also amenable to drug therapy, sometimes in conjunction with psychotherapy. The benzodiazepines continue to be widely used for the management of acute anxiety states and for rapid control of panic attacks. They are also used, though less commonly, in the long-term management of GAD and panic disorders. Anxiety symptoms may be relieved by many benzodiazepines, but it is not always easy to demonstrate the superiority of one drug over another. Alprazolam has been used in the treatment of panic disorders and agoraphobia and appears to be more selective in these conditions than other benzodiazepines. The choice of benzodiazepines for anxiety is based on several sound pharmacologic principles: (1) a rapid onset of action; (2) a relatively high therapeutic index (see drug B in Figure 22–1), plus availability of flumazenil for treatment of overdose; (3) a low risk of drug interactions based on liver enzyme induction; (4) minimal effects on cardiovascular or autonomic functions. Disadvantages of the benzodiazepines include the risk of dependence, depression of CNS functions, and amnestic effects. In addition, the benzodiazepines exert additive CNS depression when administered with other drugs, including ethanol. The patient should be warned


of this possibility to avoid impairment of performance of any task requiring mental alertness and motor coordination. In the treatment of generalized anxiety disorders and certain phobias, newer antidepressants, including selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs), are now considered by many authorities to be drugs of first choice (see Chapter 30). However, these agents have a slow onset of action and thus minimal effectiveness in acute anxiety states. Sedative-hypnotics should be used with appropriate caution so as to minimize adverse effects. A dose should be prescribed that does not impair mentation or motor functions during waking hours. Some patients may tolerate the drug better if most of the daily dose is given at bedtime, with smaller doses during the day. Prescriptions should be written for short periods, since there is little justification for longterm therapy (defined as use of therapeutic doses for 2 months or longer). The physician should make an effort to assess the efficacy of therapy from the patient’s subjective responses. Combinations of antianxiety agents should be avoided, and people taking sedatives should be cautioned about the consumption of alcohol and the concurrent use of over-the-counter medications containing antihistaminic or anticholinergic drugs (see Chapter 63).

TREATMENT OF SLEEP PROBLEMS Sleep disorders are common and often result from inadequate treatment of underlying medical conditions or psychiatric illness. True primary insomnia is rare. Nonpharmacologic therapies that are useful for sleep problems include proper diet and exercise, avoiding stimulants before retiring, ensuring a comfortable sleeping environment, and retiring at a regular time each night. In some cases, however, the patient will need and should be given a sedative-hypnotic for a limited period. It should be noted that the abrupt discontinuance of many drugs in this class can lead to rebound insomnia. Benzodiazepines can cause a dose-dependent decrease in both REM and slow-wave sleep, though to a lesser extent than the barbiturates. The newer hypnotics, zolpidem, zaleplon, and eszopiclone, are less likely than the benzodiazepines to change sleep patterns. However, so little is known about the clinical impact of these effects that statements about the desirability of a particular drug based on its effects on sleep architecture have more theoretical than practical significance. Clinical criteria of efficacy in alleviating a particular sleeping problem are more useful. The drug selected should be one that provides sleep of fairly rapid onset (decreased sleep latency) and sufficient duration, with minimal “hangover” effects such as drowsiness, dysphoria, and mental or motor depression the following day. Older drugs such as chloral hydrate, secobarbital, and pentobarbital continue to be used, but benzodiazepines, zolpidem, zaleplon, or eszopiclone are generally preferred. Daytime sedation is more common with benzodiazepines that have slow elimination rates (eg, lorazepam) and those that are biotransformed to active metabolites (eg, flurazepam, quazepam). If benzodiazepines are used nightly, tolerance can occur, which may lead to dose increases by the patient to produce the desired effect. Anterograde amnesia occurs to some degree with all benzodiazepines used for hypnosis. Eszopiclone, zaleplon, and zolpidem have efficacies similar to those of the hypnotic benzodiazepines in the management of sleep disorders. Favorable clinical features of zolpidem and the other newer hypnotics include rapid onset of activity and modest day-after psychomotor depression with few amnestic effects. Zolpidem, one of the most frequently prescribed hypnotic drugs in the United States, is available in a biphasic release formulation that provides sustained drug levels for sleep maintenance. Zaleplon acts rapidly, and because of its short half-life, the drug appears to have value in the management of patients who awaken early in the sleep cycle. At recommended doses, zaleplon and eszopiclone (despite its relatively long half-life) appear to cause less amnesia or day-after somnolence than zolpidem or benzodiazepines. The drugs in this class commonly used for sedation and hypnosis are listed in Table 22–3 together with recommended doses. Note: The failure of insomnia to remit after 7–10 days of treatment may indicate the presence of a primary psychiatric or medical illness that should be evaluated. Long-term use of hypnotics is an irrational and dangerous medical practice. TABLE 22–3 Dosages of drugs used commonly for sedation and hypnosis.


OTHER THERAPEUTIC USES Table 22–2 summarizes several other important clinical uses of drugs in the sedative-hypnotic class. Drugs used in the management of seizure disorders and as intravenous agents in anesthesia are discussed in Chapters 24 and 25. For sedative and possible amnestic effects during medical or surgical procedures such as endoscopy and bronchoscopy—as well as for premedication prior to anesthesia—oral formulations of shorter-acting drugs are preferred. Long-acting drugs such as chlordiazepoxide and diazepam and, to a lesser extent, phenobarbital are administered in progressively decreasing doses to patients during withdrawal from physiologic dependence on ethanol or other sedative-hypnotics. Parenteral lorazepam is used to suppress the symptoms of delirium tremens. Meprobamate and the benzodiazepines have frequently been used as central muscle relaxants, though evidence for general efficacy without accompanying sedation is lacking. A possible exception is diazepam, which has useful relaxant effects in skeletal muscle spasticity of central origin (see Chapter 27). Psychiatric uses of benzodiazepines other than treatment of anxiety states include the initial management of mania and the control of drug-induced hyperexcitability states (eg, phencyclidine intoxication). Sedative-hypnotics are also used occasionally as diagnostic aids in neurology and psychiatry.

CLINICAL TOXICOLOGY OF SEDATIVE-HYPNOTICS Direct Toxic Actions Many of the common adverse effects of sedative-hypnotics result from dose-related depression of the CNS. Relatively low doses may lead to drowsiness, impaired judgment, and diminished motor skills, sometimes with a significant impact on driving ability, job performance, and personal relationships. Sleep driving and other somnambulistic behavior with no memory of the event has occurred with the sedative-hypnotic drugs used in sleep disorders, prompting the FDA in 2007 to issue warnings of this potential hazard. Benzodiazepines may cause a significant dose-related anterograde amnesia; they can significantly impair ability to learn new information,


particularly that involving effortful cognitive processes, while leaving the retrieval of previously learned information intact. This effect is utilized for uncomfortable clinical procedures, eg, endoscopy, because the patient is able to cooperate during the procedure but amnesic regarding it afterward. The criminal use of benzodiazepines in cases of “date rape” is based on their dose-dependent amnestic effects. Hangover effects are not uncommon following use of hypnotic drugs with long elimination half-lives. Because elderly patients are more sensitive to the effects of sedative-hypnotics, doses approximately half of those used in younger adults are safer and usually as effective. The most common reversible cause of confusional states in the elderly is overuse of sedative-hypnotics. At higher doses, toxicity may present as lethargy or a state of exhaustion or, alternatively, as gross symptoms equivalent to those of ethanol intoxication. The physician should be aware of variability among patients in terms of doses causing adverse effects. An increased sensitivity to sedativehypnotics is more common in patients with cardiovascular disease, respiratory disease, or hepatic impairment and in older patients. Sedative-hypnotics can exacerbate breathing problems in patients with chronic pulmonary disease and in those with symptomatic sleep apnea. Sedative-hypnotics are the drugs most frequently involved in deliberate overdoses, in part because of their general availability as very commonly prescribed pharmacologic agents. The benzodiazepines are considered to be safer drugs in this respect, since they have flatter dose-response curves. Epidemiologic studies on the incidence of drug-related deaths support this general assumption—eg, 0.3 deaths per million tablets of diazepam prescribed versus 11.6 deaths per million capsules of secobarbital in one study. Alprazolam is purportedly more toxic in overdose than other benzodiazepines. Of course, many factors other than the specific sedative-hypnotic could influence such data—particularly the presence of other CNS depressants, including ethanol. In fact, most serious cases of drug overdosage, intentional or accidental, do involve polypharmacy; and when combinations of agents are taken, the practical safety of benzodiazepines may be less than the foregoing would imply. The lethal dose of any sedative-hypnotic varies with the patient and the circumstances (see Chapter 58). If discovery of the ingestion is made early and a conservative treatment regimen is started, the outcome is rarely fatal, even following very high doses. On the other hand, for most sedative-hypnotics—with the exception of benzodiazepines and possibly the newer hypnotic drugs that have a similar mechanism of action—a dose as low as ten times the hypnotic dose may be fatal if the patient is not discovered or does not seek help in time. With severe toxicity, the respiratory depression from central actions of the drug may be complicated by aspiration of gastric contents in the unattended patient—an even more likely occurrence if ethanol is present. Cardiovascular depression further complicates successful resuscitation. In such patients, treatment consists of ensuring a patent airway, with mechanical ventilation if needed, and maintenance of plasma volume, renal output, and cardiac function. Use of a positive inotropic drug such as dopamine, which preserves renal blood flow, is sometimes indicated. Hemodialysis or hemoperfusion may be used to hasten elimination of some of these drugs (see Table 58-3). Flumazenil reverses the sedative actions of benzodiazepines, and those of eszopiclone, zaleplon, and zolpidem, although experience with its use in overdose of the newer hypnotics is limited. However, its duration of action is short, its antagonism of respiratory depression is unpredictable, and there is a risk of precipitation of withdrawal symptoms in long-term users of benzodiazepines. Consequently, the use of flumazenil in benzodiazepine overdose remains controversial and must be accompanied by adequate monitoring and support of respiratory function. The extensive clinical use of triazolam has led to reports of serious CNS effects including behavioral disinhibition, delirium, aggression, and violence. However, behavioral disinhibition may occur with any sedative-hypnotic drug, and it does not appear to be more prevalent with triazolam than with other benzodiazepines. Disinhibitory reactions during benzodiazepine treatment are more clearly associated with the use of very high doses and the pretreatment level of patient hostility. Adverse effects of the sedative-hypnotics that are not referable to their CNS actions occur infrequently. Hypersensitivity reactions, including skin rashes, occur only occasionally with most drugs of this class. Reports of teratogenicity leading to fetal deformation following use of certain benzodiazepines have resulted in FDA assignment of individual benzodiazepines to either category D or X in terms of pregnancy risk. Most barbiturates are FDA pregnancy category D. Eszopiclone, ramelteon, zaleplon, and zolpidem are category C, while buspirone is a category B drug in terms of use in pregnancy. Because barbiturates enhance porphyrin synthesis, they are absolutely contraindicated in patients with a history of acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, or symptomatic porphyria.

Alterations in Drug Response Depending on the dosage and the duration of use, tolerance occurs in varying degrees to many of the pharmacologic effects of sedativehypnotics. However, it should not be assumed that the degree of tolerance achieved is identical for all pharmacologic effects. There is evidence that the lethal dose range is not altered significantly by the long-term use of sedative-hypnotics. Cross-tolerance between the different sedative-hypnotics, including ethanol, can lead to an unsatisfactory therapeutic response when standard doses of a drug are used in a patient with a recent history of excessive use of these agents. However, there have been very few reports of tolerance development when eszopiclone, zolpidem, or zaleplon was used for less than 4 weeks. With the long-term use of sedative-hypnotics, especially if doses are increased, a state of physiologic dependence can occur. This may develop to a degree unparalleled by any other drug group, including the opioids. Withdrawal from a sedative-hypnotic can have severe and life-threatening manifestations. Withdrawal symptoms range from restlessness, anxiety, weakness, and orthostatic hypotension to hyperactive reflexes and generalized seizures. Symptoms of withdrawal are usually more severe following discontinuance


of sedative-hypnotics with shorter half-lives. However, eszopiclone, zolpidem, and zaleplon appear to be exceptions to this, because withdrawal symptoms are minimal following abrupt discontinuance of these newer short-acting agents. Symptoms are less pronounced with longer-acting drugs, which may partly accomplish their own “tapered� withdrawal by virtue of their slow elimination. Crossdependence, defined as the ability of one drug to suppress abstinence symptoms from discontinuance of another drug, is quite marked among sedative-hypnotics. This provides the rationale for therapeutic regimens in the management of withdrawal states: Longer-acting drugs such as chlordiazepoxide, diazepam, and phenobarbital can be used to alleviate withdrawal symptoms of shorter-acting drugs, including ethanol.

Drug Interactions The most common drug interactions involving sedative-hypnotics are interactions with other CNS depressant drugs, leading to additive effects. These interactions have some therapeutic usefulness when these drugs are used as adjuvants in anesthesia practice. However, if not anticipated, such interactions can lead to serious consequences, including enhanced depression with concomitant use of many other drugs. Additive effects can be predicted with concomitant use of alcoholic beverages, opioid analgesics, anticonvulsants, and phenothiazines. Less obvious but just as important is enhanced CNS depression with a variety of antihistamines, antihypertensive agents, and antidepressant drugs of the tricyclic class. Interactions involving changes in the activity of hepatic drug-metabolizing enzyme systems have been discussed (see also Chapters 4 and 66).

SUMMARY Sedative-Hypnotics



PREPARATIONS AVAILABLE

REFERENCES Ancoli-Israel S et al: Long-term use of sedative hypnotics in older patients with insomnia. Sleep Med 2005;6:107. Bateson AN: T he benzodiazepine site of the GABA A receptor: An old target with new potential? Sleep Med 2004;5(Suppl 1):S9. Chouinard G: Issues in the clinical use of benzodiazepines: Potency, withdrawal, and rebound. J Clin Psychiatry 2004;65(Suppl 5):7. Clayton T et al: An updated unified pharmacophore model of the benzodiazepine binding site on gamma-aminobutyric acid(a) receptors: Correlation with comparative models. Curr Med Chem 2007;14:2755. Cloos JM, Ferreira V: Current use of benzodiazepines in anxiety disorders. Curr Opin Psychiatry 2009;22:90. Da Settimo F et al: GABA A/Bz receptor subtypes as targets for selective drugs. Curr Med Chem 2007;14:2680. Davidson JR et al: A psychopharmacological treatment algorithm for generalized anxiety disorder. J Psychopharmacol 2010;24:3. Drover DR: Comparative pharmacokinetics and pharmacodynamics of short-acting hypnosedatives: Zaleplon, zolpidem and zopiclone. Clin Pharmacokinet 2004;43:227. Drugs for Insomnia. T reatment Guidelines 2012;10:57. Erman M et al: An efficacy, safety, and dose-response study of ramelteon in patients with chronic primary insomnia. Sleep Med 2006;7:17. Gottesmann C: GABA mechanisms and sleep. Neuroscience 2002; 111:231.


Hanson SM, Czajkowski C: Structural mechanisms underlying benzodiazepine modulation of the GABA(A) receptor. J Neurosci 2008;28:3490. Hesse LM et al: Clinically important drug interactions with zopiclone, zolpidem and zaleplon. CNS Drugs 2003;17:513. Kato K et al: Neurochemical properties of ramelteon, a selective MT 1/MT 2 receptor agonist. Neuropharmacology 2005;48:301. Kralic JE et al: GABA(A) receptor alpha-1 subunit deletion alters receptor subtype assembly, pharmacological and behavioral responses to benzodiazepines and zolpidem. Neuropharmacology 2002;43:685. Krystal AD: T he changing perspective of chronic insomnia management. J Clin Psychiatry 2004;65(Suppl 8):20. Lader M, T ylee A, Donoghue J: Withdrawing benzodiazepines in primary care. CNS Drugs 2009;23:2319. McKernan RM et al: Anxiolytic-like action of diazepam: Which GABA(A) receptor subtype is involved? T rends Pharmacol Sci 2001;22:402. Mohler H, Fritschy JM, Rudolph U: A new benzodiazepine pharmacology. J Pharmacol Exp T her 2002;300:2. Morairty SR, et al: T he hypocretin/orexin antagonist almorexant promotes sleep without impairment of performance in rats. Front Neurosci 2014;8:3. Neubauer DN: New directions in the pharmacologic treatment of insomnia. Primary Psychiatry 2006;13:51. Rapaport MJ et al: Benzodiazepine use and driving: A meta analysis. J Clin Psychiatry 2009;70:663. Rosenberg R et al: An assessment of the efficacy and safety of eszopiclone in the treatment of transient insomnia in healthy adults. Sleep Med 2005;6:15. Sanger DJ: T he pharmacology and mechanism of action of new generation, non-benzodiazepine hypnotic agents. CNS Drugs 2004; 18(Suppl 1):9. Silber MH: Chronic insomnia. N Engl J Med 2005;353:803. Walsh JK: Pharmacologic management of insomnia. J Clin Psychiatry 2004;65(Suppl 16):41. Wurtman R: Ramelteon: A novel treatment for the treatment of insomnia. Expert Rev Neurother 2006;6:957. *

In memory of Walter (Skip) Way, MD, with thanks for his past contributions to this chapter.

CASE STUDY ANSWER As described in this chapter, nonpharmacologic factors are very important in the management of sleep problems: proper diet (and avoidance of snacks before bedtime), exercise, and a regular time and place for sleep. Avoidance of stimulants is very important, and the large intake of diet colas reported by the patient should be reduced, especially in the latter half of the day. If problems persist after these measures are implemented, one of the newer hypnotics (eszopiclone, zaleplon, or zolpidem) may be tried on a short-term basis.


CHAPTER

23 The Alcohols Susan B. Masters, PhD, & Anthony J. Trevor, PhD

CASE STUDY An 18-year-old college freshman began drinking alcohol at 8:30 PM during a hazing event at his new fraternity. Between 8:30 and approximately midnight, he and several other pledges consumed beer and a bottle of whiskey, and then he consumed most of a bottle of rum at the urging of upperclassmen. The young man complained of feeling nauseated, lay down on a couch, and began to lose consciousness. Two upperclassmen carried him to his bedroom, placed him on his stomach, and positioned a trash can nearby. Approximately 10 minutes later, the freshman was found unconscious and covered with vomit. There was a delay in treatment because the upperclassmen called the college police instead of calling 911. After the call was transferred to 911, emergency medical technicians responded quickly and discovered that the young man was not breathing and that he had choked on his vomit. He was rushed to the hospital, where he remained in a coma for 2 days before ultimately being pronounced dead. The patient’s blood alcohol concentration shortly after arriving at the hospital was 510 mg/dL. What was the cause of this patient’s death? If he had received medical care sooner, what treatment might have prevented his death?

Alcohol, primarily in the form of ethyl alcohol (ethanol), has occupied an important place in the history of humankind for at least 8000 years. In Western society, beer and wine were a main staple of daily life until the 19th century. These relatively dilute alcoholic beverages were preferred over water, which was known—long before the discovery of microbes—to be associated with acute and chronic illness. Partially sterilized by the fermentation process and the alcohol content, alcoholic beverages provided important calories and nutrients and served as a main source of daily liquid intake. As systems for improved sanitation and water purification were introduced in the 1800s, beer and wine became less important components of the human diet, and the consumption of alcoholic beverages, including distilled preparations with higher concentrations of alcohol, shifted toward their present-day role, in many societies, as a socially acceptable form of recreation. Today, alcohol is widely consumed. Like other sedative-hypnotic drugs, alcohol in low to moderate amounts relieves anxiety and fosters a feeling of well-being or even euphoria. However, alcohol is also the most commonly abused drug in the world, and the cause of vast medical and societal costs. In the United States, approximately 75% of the adult population drinks alcohol regularly. The majority of this drinking population is able to enjoy the pleasurable effects of alcohol without allowing alcohol consumption to become a health risk. However, about 8% of the general population in the United States has an alcohol-use disorder. Individuals who use alcohol in dangerous situations (eg, drinking and driving or combining alcohol with other medications) or continue to drink alcohol in spite of adverse consequences related directly to their alcohol consumption suffer from alcohol abuse (see also Chapter 32). Individuals with alcohol dependence have characteristics of alcohol abuse and additionally exhibit physical dependence on alcohol (tolerance to alcohol and signs and symptoms upon withdrawal). They also demonstrate an inability to control their drinking and devote much time to getting and using alcohol, or recovering from its effects. The alcohol-use disorders are complex, with genetic as well as environmental determinants. The societal and medical costs of alcohol abuse are staggering. It is estimated that about 30% of all people admitted to hospitals have coexisting alcohol problems. Once in the hospital, people with chronic alcoholism generally have poorer outcomes. In addition, each year tens of thousands of children are born with morphologic and functional defects resulting from prenatal exposure to ethanol. Despite the investment of many resources and much basic research, alcoholism remains a common chronic disease that is difficult to treat. Ethanol and many other alcohols with potentially toxic effects are used as fuels and in industry—some in enormous quantities. In addition to ethanol, methanol and ethylene glycol toxicity occurs with sufficient frequency to warrant discussion in this chapter.

BASIC PHARMACOLOGY OF ETHANOL


Pharmacokinetics Ethanol is a small water-soluble molecule that is absorbed rapidly from the gastrointestinal tract. After ingestion of alcohol in the fasting state, peak blood alcohol concentrations are reached within 30 minutes. The presence of food in the stomach delays absorption by slowing gastric emptying. Distribution is rapid, with tissue levels approximating the concentration in blood. The volume of distribution for ethanol approximates total body water (0.5–0.7 L/kg). After an equivalent oral dose of alcohol, women have a higher peak concentration than men, in part because women have a lower total body water content and in part because of differences in first-pass metabolism. In the central nervous system (CNS), the concentration of ethanol rises quickly, since the brain receives a large proportion of total blood flow and ethanol readily crosses biologic membranes. Over 90% of alcohol consumed is oxidized in the liver; much of the remainder is excreted through the lungs and in the urine. The excretion of a small but consistent proportion of alcohol by the lungs can be quantified with breath alcohol tests that serve as a basis for a legal definition of “driving under the influence (DUI)” in many countries. At levels of ethanol usually achieved in blood, the rate of oxidation follows zero-order kinetics; that is, it is independent of time and concentration of the drug. The typical adult can metabolize 7– 10 g (150–220 mmol) of alcohol per hour, the equivalent of approximately one “drink” [10 oz (300 mL) beer, 3.5 oz (105 mL) wine, or 1 oz (30 mL) distilled 80-proof spirits]. Two major pathways of alcohol metabolism to acetaldehyde have been identified (Figure 23–1). Acetaldehyde is then oxidized to acetate by a third metabolic process.

FIGURE 23–1 Metabolism of ethanol by alcohol dehydrogenase and the microsomal ethanol-oxidizing system (MEOS). Alcohol dehydrogenase and aldehyde dehydrogenase are inhibited by fomepizole and disulfiram, respectively. NAD+, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate. A. Alcohol Dehydrogenase Pathway The primary pathway for alcohol metabolism involves alcohol dehydrogenase (ADH), a family of cytosolic enzymes that catalyze the conversion of alcohol to acetaldehyde (Figure 23–1, left). These enzymes are located mainly in the liver, but small amounts are found in other organs such as the brain and stomach. There is considerable genetic variation in ADH enzymes, affecting the rate of ethanol metabolism and also appearing to alter vulnerability to alcohol-abuse disorders. For example, one ADH allele (the ADH1B*2 allele), which is associated with rapid conversion of ethanol to acetaldehyde, has been found to be protective against alcohol dependence in several ethnic populations and especially East Asians.


Some metabolism of ethanol by ADH occurs in the stomach in men, but a smaller amount occurs in women, who appear to have lower levels of the gastric enzyme. This difference in gastric metabolism of alcohol in women probably contributes to the sex-related differences in blood alcohol concentrations noted above. During conversion of ethanol by ADH to acetaldehyde, hydrogen ion is transferred from ethanol to the cofactor nicotinamide adenine dinucleotide (NAD+) to form NADH. As a net result, alcohol oxidation generates an excess of reducing equivalents in the liver, chiefly as NADH. The excess NADH production appears to contribute to the metabolic disorders that accompany chronic alcoholism and to both the lactic acidosis and hypoglycemia that frequently accompany acute alcohol poisoning. B. Microsomal Ethanol-Oxidizing System (MEOS) This enzyme system, also known as the mixed function oxidase system, uses NADPH as a cofactor in the metabolism of ethanol (Figure 23–1, right) and consists primarily of cytochrome P450 2E1, 1A2, and 3A4 (see Chapter 4). During chronic alcohol consumption, MEOS activity is induced. As a result, chronic alcohol consumption results in significant increases not only in ethanol metabolism but also in the clearance of other drugs eliminated by the cytochrome P450s that constitute the MEOS system, and in the generation of the toxic byproducts of cytochrome P450 reactions (toxins, free radicals, H2 O2 ). C. Acetaldehyde Metabolism Much of the acetaldehyde formed from alcohol is oxidized in the liver in a reaction catalyzed by mitochondrial NAD-dependent aldehyde dehydrogenase (ALDH). The product of this reaction is acetate (Figure 23–1), which can be further metabolized to CO2 and water, or used to form acetyl-CoA. Oxidation of acetaldehyde is inhibited by disulfiram, a drug that has been used to deter drinking by patients with alcohol dependence. When ethanol is consumed in the presence of disulfiram, acetaldehyde accumulates and causes an unpleasant reaction of facial flushing, nausea, vomiting, dizziness, and headache. Several other drugs (eg, metronidazole, cefotetan, trimethoprim) inhibit ALDH and can cause a disulfiram-like reaction if combined with ethanol. Some people, primarily of East Asian descent, have genetic deficiency in the activity of the mitochondrial form of ALDH, which is encoded by the ALDH2 gene. When these individuals drink alcohol, they develop high blood acetaldehyde concentrations and experience a noxious reaction similar to that seen with the combination of disulfiram and ethanol. This form of reduced-activity ALDH is strongly protective against alcohol-use disorders.

Pharmacodynamics of Acute Ethanol Consumption A. Central Nervous System The CNS is markedly affected by acute alcohol consumption. Alcohol causes sedation, relief of anxiety and, at higher concentrations, slurred speech, ataxia, impaired judgment, and disinhibited behavior, a condition usually called intoxication or drunkenness (Table 23–1). These CNS effects are most marked as the blood level is rising, because acute tolerance to the effects of alcohol occurs after a few hours of drinking. For chronic drinkers who are tolerant to the effects of alcohol, higher concentrations are needed to elicit these CNS effects. For example, an individual with chronic alcoholism may appear sober or only slightly intoxicated with a blood alcohol concentration of 300–400 mg/dL, whereas this level is associated with marked intoxication or even coma in a nontolerant individual. The propensity of moderate doses of alcohol to inhibit the attention and information-processing skills as well as the motor skills required for operation of motor vehicles has profound effects. Approximately 30–40% of all traffic accidents resulting in a fatality in the United States involve at least one person with blood alcohol near or above the legal level of intoxication, and drunken driving is a leading cause of death in young adults. TABLE 23–1 Blood alcohol concentration (BAC) and clinical effects in nontolerant individuals.


Like other sedative-hypnotic drugs, alcohol is a CNS depressant. At high blood concentrations, it induces coma, respiratory depression, and death. Ethanol affects a large number of membrane proteins that participate in signaling pathways, including neurotransmitter receptors for amines, amino acids, opioids, and neuropeptides; enzymes such as Na+/K+-ATPase, adenylyl cyclase, phosphoinositide-specific phospholipase C; a nucleoside transporter; and ion channels. Much attention has focused on alcohol’s effects on neurotransmission by glutamate and γ-aminobutyric acid (GABA), the main excitatory and inhibitory neurotransmitters in the CNS. Acute ethanol exposure enhances the action of GABA at GABA A receptors, which is consistent with the ability of GABA-mimetics to intensify many of the acute effects of alcohol and of GABAA antagonists to attenuate some of the actions of ethanol. Ethanol inhibits the ability of glutamate to open the cation channel associated with the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors. The NMDA receptor is implicated in many aspects of cognitive function, including learning and memory. “Blackouts”—periods of memory loss that occur with high levels of alcohol—may result from inhibition of NMDA receptor activation. Experiments that use modern genetic approaches eventually will yield a more precise definition of ethanol’s direct and indirect targets. In recent years, experiments with mutant strains of mice, worms, and flies have reinforced the importance of previously identified targets and helped identify new candidates, including a calcium-regulated and voltage-gated potassium channel that may be one of ethanol’s direct targets (see Box: What Can Drunken Worms, Flies, and Mice Tell Us about Alcohol?). B. Heart Significant depression of myocardial contractility has been observed in individuals who acutely consume moderate amounts of alcohol, ie, at a blood concentration above 100 mg/dL. C. Smooth Muscle Ethanol is a vasodilator, probably as a result of both CNS effects (depression of the vasomotor center) and direct smooth muscle relaxation caused by its metabolite, acetaldehyde. In cases of severe overdose, hypothermia—caused by vasodilation—may be marked in cold environments. Ethanol also relaxes the uterus and—before the introduction of more effective and safer uterine relaxants (eg, calcium channel antagonists)—was used intravenously for the suppression of premature labor.

Consequences of Chronic Alcohol Consumption Chronic alcohol consumption profoundly affects the function of several vital organs—particularly the liver—and the nervous, gastrointestinal, cardiovascular, and immune systems. Since ethanol has low potency, it requires concentrations thousands of times higher than other misused drugs (eg, cocaine, opiates, amphetamines) to produce its intoxicating effects. As a result, ethanol is consumed in quantities that are unusually large for a pharmacologically active drug. The tissue damage caused by chronic alcohol ingestion results from a combination of the direct effects of ethanol and acetaldehyde, and the metabolic consequences of processing a heavy load of a


metabolically active substance. Specific mechanisms implicated in tissue damage include increased oxidative stress coupled with depletion of glutathione, damage to mitochondria, growth factor dysregulation, and potentiation of cytokine-induced injury.

What Can Drunken Worms, Flies, and Mice Tell Us about Alcohol? For a drug like ethanol, which exhibits low potency and specificity, and modifies complex behaviors, the precise roles of its many direct and indirect targets are difficult to define. Increasingly, ethanol researchers are employing genetic approaches to complement standard neurobiologic experimentation. Three experimental animal systems for which powerful genetic techniques exist—mice, flies, and worms—have yielded intriguing results. Strains of mice with abnormal sensitivity to ethanol were identified many years ago by breeding and selection programs. Using sophisticated genetic mapping and sequencing techniques, researchers have made progress in identifying the genes that confer these traits. A more targeted approach is the use of transgenic mice to test hypotheses about specific genes. For example, after earlier experiments suggested a link between brain neuropeptide Y (NPY) and ethanol, researchers used two transgenic mouse models to further investigate the link. They found that a strain of mice that lacks the gene for NPY—NPY knockout mice—consume more ethanol than control mice and are less sensitive to ethanol’s sedative effects. As would be expected if increased concentrations of NPY in the brain make mice more sensitive to ethanol, a strain of mice that overexpresses NPY drinks less alcohol than the controls even though their total consumption of food and liquid is normal. Work with other transgenic knockout mice supports the central role in ethanol responses of signaling systems that have long been believed to be involved (eg, GABAA, glutamate, dopamine, opioid, and serotonin receptors) and has helped build the case for newer candidates such as NPY and corticotropinreleasing hormone, cannabinoid receptors, ion channels, and protein kinase C. It is easy to imagine mice having measurable behavioral responses to alcohol, but drunken worms and fruit flies are harder to imagine. Actually, both invertebrates respond to ethanol in ways that parallel mammalian responses. Drosophila melanogaster fruit flies exposed to ethanol vapor show increased locomotion at low concentrations but at higher concentrations, become poorly coordinated, sedated, and finally immobile. These behaviors can be monitored by sophisticated laser or video tracking methods or with an ingenious “chromatography” column of air that separates relatively insensitive flies, from inebriated flies, which drop to the bottom of the column. The worm Caenorhabditis elegans similarly exhibits increased locomotion at low ethanol concentrations and, at higher concentrations, reduced locomotion, sedation, and—something that can be turned into an effective screen for mutant worms that are resistant to ethanol—impaired egg laying. The advantage of using flies and worms as genetic models for ethanol research is their relatively simple neuroanatomy, well-established techniques for genetic manipulation, an extensive library of wellcharacterized mutants, and completely or nearly completely solved genetic codes. Already, much information has accumulated about candidate proteins involved with the effects of ethanol in flies. In an elegant study on C elegans, researchers found evidence that a calcium-activated, voltage-gated BK potassium channel is a direct target of ethanol. This channel, which is activated by ethanol, has close homologs in flies and vertebrates, and evidence is accumulating that ethanol has similar effects in these homologs. Genetic experiments in these model systems should provide information that will help narrow and focus research into the complex and important effects of ethanol in humans. Chronic consumption of large amounts of alcohol is associated with an increased risk of death. Deaths linked to alcohol consumption are caused by liver disease, cancer, accidents, and suicide. A. Liver and Gastrointestinal Tract Liver disease is the most common medical complication of alcohol abuse; an estimated 15–30% of chronic heavy drinkers eventually develop severe liver disease. Alcoholic fatty liver, a reversible condition, may progress to alcoholic hepatitis and finally to cirrhosis and liver failure. In the United States, chronic alcohol abuse is the leading cause of liver cirrhosis and of the need for liver transplantation. The risk of developing liver disease is related both to the average amount of daily consumption and to the duration of alcohol abuse. Women appear to be more susceptible to alcohol hepatotoxicity than men. Concurrent infection with hepatitis B or C virus increases the risk of severe liver disease. The pathogenesis of alcoholic liver disease is a multifactorial process involving metabolic repercussions of ethanol oxidation in the liver, dysregulation of fatty acid oxidation and synthesis, and activation of the innate immune system by a combination of direct effects of ethanol and its metabolites and by bacterial endotoxins that access the liver as a result of ethanol-induced changes in the intestinal tract. Tumor necrosis factor-α, a proinflammatory cytokine that is consistently elevated in animal models of alcoholic liver disease and in patients with alcoholic liver disease, appears to play a pivotal role in the progression of alcoholic liver disease and may be a fruitful therapeutic target. Other portions of the gastrointestinal tract can also be injured. Chronic alcohol ingestion is by far the most common cause of chronic pancreatitis in the Western world. In addition to its direct toxic effect on pancreatic acinar cells, alcohol alters pancreatic epithelial permeability and promotes the formation of protein plugs and calcium carbonate-containing stones. Individuals with chronic alcoholism are prone to gastritis and have increased susceptibility to blood and plasma protein loss during


drinking, which may contribute to anemia and protein malnutrition. Alcohol also injures the small intestine, leading to diarrhea, weight loss, and multiple vitamin deficiencies. Malnutrition from dietary deficiency and vitamin deficiencies due to malabsorption are common in alcoholism. Malabsorption of water-soluble vitamins is especially severe. B. Nervous System 1. Tolerance and dependence—The consumption of alcohol in high doses over a long period results in tolerance and in physical and psychological dependence. Tolerance to the intoxicating effects of alcohol is a complex process involving poorly understood changes in the nervous system as well as the metabolic changes described earlier. As with other sedative-hypnotic drugs, there is a limit to tolerance, so that only a relatively small increase in the lethal dose occurs with increasing alcohol use. Chronic alcohol drinkers, when forced to reduce or discontinue alcohol, experience a withdrawal syndrome, which indicates the existence of physical dependence. Alcohol withdrawal symptoms usually consist of hyperexcitability in mild cases and seizures, toxic psychosis, and delirium tremens in severe ones. The dose, rate, and duration of alcohol consumption determine the intensity of the withdrawal syndrome. When consumption has been very high, merely reducing the rate of consumption may lead to signs of withdrawal. Psychological dependence on alcohol is characterized by a compulsive desire to experience the rewarding effects of alcohol and, for current drinkers, a desire to avoid the negative consequences of withdrawal. People who have recovered from alcoholism and become abstinent still experience periods of intense craving for alcohol that can be triggered by environmental cues associated in the past with drinking, such as familiar places, groups of people, or events. The molecular basis of alcohol tolerance and dependence is not known with certainty, nor is it known whether the two phenomena reflect opposing effects on a shared molecular pathway. Tolerance may result from ethanol-induced up-regulation of a pathway in response to the continuous presence of ethanol. Dependence may result from overactivity of that same pathway after the ethanol effect dissipates and before the system has time to return to a normal ethanol-free state. Chronic exposure of animals or cultured cells to alcohol elicits a multitude of adaptive responses involving neurotransmitters and their receptors, ion channels, and enzymes that participate in signal transduction pathways. Up-regulation of the NMDA subtype of glutamate receptors and voltage-sensitive Ca2+ channels may underlie the seizures that accompany the alcohol withdrawal syndrome. Based on the ability of sedative-hypnotic drugs that enhance GABAergic neurotransmission to substitute for alcohol during alcohol withdrawal and evidence of down-regulation of GABAA-mediated responses with chronic alcohol exposure, changes in GABA neurotransmission are believed to play a central role in tolerance and withdrawal. Like other drugs of abuse, ethanol modulates neural activity in the brain’s mesolimbic dopamine reward circuit and increases dopamine release in the nucleus accumbens (see Chapter 32). Alcohol affects local concentrations of serotonin, opioids, and dopamine— neurotransmitters involved in the brain reward system—and has complex effects on the expression of receptors for these neurotransmitters and their signaling pathways. The discovery that naltrexone, a nonselective opioid receptor antagonist, helps patients who are recovering from alcoholism abstain from drinking supports the idea that a common neurochemical reward system is shared by very different drugs associated with physical and psychological dependence. There is also convincing evidence from animal models that ethanol intake and seeking behavior are reduced by antagonists of another important regulator of the brain reward system, the cannabinoid CB1 receptor, which is the molecular target of active ingredients in marijuana. Two other important neuroendocrine systems that appear to play key roles in modulating ethanol-seeking activity in experimental animals are the appetite-regulating system—which uses peptides such as leptin, ghrelin, and neuropeptide Y—and the stress response system, which is controlled by corticotropin-releasing factor. 2. Neurotoxicity—Consumption of large amounts of alcohol over extended periods (usually years) often leads to neurologic deficits. The most common neurologic abnormality in chronic alcoholism is generalized symmetric peripheral nerve injury, which begins with distal paresthesias of the hands and feet. Degenerative changes can also result in gait disturbances and ataxia. Other neurologic disturbances associated with alcoholism include dementia and, rarely, demyelinating disease. Wernicke-Korsakoff syndrome is a relatively uncommon but important entity characterized by paralysis of the external eye muscles, ataxia, and a confused state that can progress to coma and death. It is associated with thiamine deficiency but is rarely seen in the absence of alcoholism. Because of the importance of thiamine in this pathologic condition and the absence of toxicity associated with thiamine administration, all patients suspected of having Wernicke-Korsakoff syndrome (including virtually all patients who present to the emergency department with altered consciousness, seizures, or both) should receive thiamine therapy. Often, the ocular signs, ataxia, and confusion improve promptly upon administration of thiamine. However, most patients are left with a chronic disabling memory disorder known as Korsakoff’s psychosis. Alcohol may also impair visual acuity, with painless blurring that occurs over several weeks of heavy alcohol consumption. Changes are usually bilateral and symmetric and may be followed by optic nerve degeneration. Ingestion of ethanol substitutes such as methanol (see Pharmacology of Other Alcohols) causes severe visual disturbances. C. Cardiovascular System


1. Cardiomyopathy and heart failure—Alcohol has complex effects on the cardiovascular system. Heavy alcohol consumption of long duration is associated with a dilated cardiomyopathy with ventricular hypertrophy and fibrosis. In animals and humans, alcohol induces a number of changes in heart cells that may contribute to cardiomyopathy. They include membrane disruption, depressed function of mitochondria and sarcoplasmic reticulum, intracellular accumulation of phospholipids and fatty acids, and up-regulation of voltage-gated calcium channels. There is evidence that patients with alcohol-induced dilated cardiomyopathy do significantly worse than patients with idiopathic dilated cardiomyopathy, even though cessation of drinking is associated with a reduction in cardiac size and improved function. The poorer prognosis for patients who continue to drink appears to be due in part to interference by ethanol with the beneficial effects of β blockers and angiotensin-converting enzyme (ACE) inhibitors. 2. Arrhythmias—Heavy drinking—and especially “binge” drinking—are associated with both atrial and ventricular arrhythmias. Patients undergoing alcohol withdrawal syndrome can develop severe arrhythmias that may reflect abnormalities of potassium or magnesium metabolism as well as enhanced release of catecholamines. Seizures, syncope, and sudden death during alcohol withdrawal may be due to these arrhythmias. 3. Hypertension—A link between heavier alcohol consumption (more than three drinks per day) and hypertension has been firmly established in epidemiologic studies. Alcohol is estimated to be responsible for approximately 5% of cases of hypertension, making it one of the most common causes of reversible hypertension. This association is independent of obesity, salt intake, coffee drinking, and cigarette smoking. A reduction in alcohol intake appears to be effective in lowering blood pressure in hypertensive individuals who are also heavy drinkers; the hypertension seen in this population is also responsive to standard blood pressure medications. 4. Coronary heart disease—Although the deleterious effects of excessive alcohol use on the cardiovascular system are well established, there is strong epidemiologic evidence that moderate alcohol consumption actually prevents coronary heart disease (CHD), ischemic stroke, and peripheral arterial disease. This type of relationship between mortality and the dose of a drug is called a “J-shaped” relationship. Results of these clinical studies are supported by ethanol’s ability to raise serum levels of high-density lipoprotein (HDL) cholesterol (the form of cholesterol that appears to protect against atherosclerosis; see Chapter 35), by its ability to inhibit some of the inflammatory processes that underlie atherosclerosis while also increasing production of the endogenous anticoagulant tissue plasminogen activator (t-PA, see Chapter 34), and by the presence in alcoholic beverages (especially red wine) of antioxidants and other substances that may protect against atherosclerosis. These observational studies are intriguing, but randomized clinical trials examining the possible benefit of moderate alcohol consumption in prevention of CHD have not been carried out. D. Blood Alcohol indirectly affects hematopoiesis through metabolic and nutritional effects and may also directly inhibit the proliferation of all cellular elements in bone marrow. The most common hematologic disorder seen in chronic drinkers is mild anemia resulting from alcoholrelated folic acid deficiency. Iron deficiency anemia may result from gastrointestinal bleeding. Alcohol has also been implicated as a cause of several hemolytic syndromes, some of which are associated with hyperlipidemia and severe liver disease. E. Endocrine System and Electrolyte Balance Chronic alcohol use has important effects on the endocrine system and on fluid and electrolyte balance. Clinical reports of gynecomastia and testicular atrophy in alcoholics with or without cirrhosis suggest a derangement in steroid hormone balance. Individuals with chronic liver disease may have disorders of fluid and electrolyte balance, including ascites, edema, and effusions. Alterations of whole body potassium induced by vomiting and diarrhea, as well as severe secondary aldosteronism, may contribute to muscle weakness and can be worsened by diuretic therapy. The metabolic derangements caused by metabolism of large amounts of ethanol can result in hypoglycemia, as a result of impaired hepatic gluconeogenesis, and in ketosis, caused by excessive lipolytic factors, especially increased cortisol and growth hormone. F. Fetal Alcohol Syndrome Chronic maternal alcohol abuse during pregnancy is associated with teratogenic effects, and alcohol is a leading cause of mental retardation and congenital malformation. The abnormalities that have been characterized as fetal alcohol syndrome include (1) intrauterine growth retardation, (2) microcephaly, (3) poor coordination, (4) underdevelopment of midfacial region (appearing as a flattened face), and (5) minor joint anomalies. More severe cases may include congenital heart defects and mental retardation. Although the level of alcohol intake required to cause serious neurologic deficits appears quite high, the threshold for more subtle neurologic deficits is uncertain. The mechanisms that underlie ethanol’s teratogenic effects are unknown. Ethanol rapidly crosses the placenta and reaches concentrations in the fetus that are similar to those in maternal blood. The fetal liver has little or no alcohol dehydrogenase activity, so the fetus must rely on maternal and placental enzymes for elimination of alcohol. The neuropathologic abnormalities seen in humans and in animal models of fetal alcohol syndrome indicate that ethanol triggers apoptotic neurodegeneration and also causes aberrant neuronal and glial migration in the developing nervous system. In tissue culture


systems, ethanol causes a striking reduction in neurite outgrowth. G. Immune System The effects of alcohol on the immune system are complex; immune function in some tissues is inhibited (eg, the lung), whereas pathologic, hyperactive immune function in other tissues is triggered (eg, liver, pancreas). In addition, acute and chronic exposure to alcohol have widely different effects on immune function. The types of immunologic changes reported for the lung include suppression of the function of alveolar macrophages, inhibition of chemotaxis of granulocytes, and reduced number and function of T cells. In the liver, there is enhanced function of key cells of the innate immune system (eg, Kupffer cells, hepatic stellate cells) and increased cytokine production. In addition to the inflammatory damage that chronic heavy alcohol use precipitates in the liver and pancreas, it predisposes to infections, especially of the lung, and worsens the morbidity and increases the mortality risk of patients with pneumonia. H. Increased Risk of Cancer Chronic alcohol use increases the risk for cancer of the mouth, pharynx, larynx, esophagus, and liver. Evidence also points to a small increase in the risk of breast cancer in women. Much more information is required before a threshold level for alcohol consumption as it relates to cancer can be established. Alcohol itself does not appear to be a carcinogen in most test systems. However, its primary metabolite, acetaldehyde, can damage DNA, as can the reactive oxygen species produced by increased cytochrome P450 activity. Other factors implicated in the link between alcohol and cancer include changes in folate metabolism and the growth-promoting effects of chronic inflammation.

Alcohol-Drug Interactions Interactions between ethanol and other drugs can have important clinical effects resulting from alterations in the pharmacokinetics or pharmacodynamics of the second drug. The most common pharmacokinetic alcohol-drug interactions stem from alcohol-induced increases of drug-metabolizing enzymes, as described in Chapter 4. Thus, prolonged intake of alcohol without damage to the liver can enhance the metabolic biotransformation of other drugs. Ethanol-mediated induction of hepatic cytochrome P450 enzymes is particularly important with regard to acetaminophen. Chronic consumption of three or more drinks per day increases the risk of hepatotoxicity due to toxic or even high therapeutic levels of acetaminophen as a result of increased P450-mediated conversion of acetaminophen to reactive hepatotoxic metabolites (see Figure 4– 5). Current FDA regulations require that over-the-counter products containing acetaminophen carry a warning about the relation between ethanol consumption and acetaminophen-induced hepatotoxicity. In contrast, acute alcohol use can inhibit metabolism of other drugs because of decreased enzyme activity or decreased liver blood flow. Phenothiazines, tricyclic antidepressants, and sedative-hypnotic drugs are the most important drugs that interact with alcohol by this pharmacokinetic mechanism. Pharmacodynamic interactions are also of great clinical significance. The additive CNS depression that occurs when alcohol is combined with other CNS depressants, particularly sedative-hypnotics, is most important. Alcohol also potentiates the pharmacologic effects of many nonsedative drugs, including vasodilators and oral hypoglycemic agents.

CLINICAL PHARMACOLOGY OF ETHANOL Alcohol is the cause of more preventable morbidity and mortality than all other drugs combined with the exception of tobacco. The search for specific etiologic factors or the identification of significant predisposing variables for alcohol abuse has led to disappointing results. Personality type, severe life stresses, psychiatric disorders, and parental role models are not reliable predictors of alcohol abuse. Although environmental factors clearly play a role, evidence suggests that there is a large genetic contribution to the development of alcoholism. Not surprisingly, polymorphisms in alcohol dehydrogenase and aldehyde dehydrogenase that lead to increased aldehyde accumulation and its associated facial flushing, nausea, and hypotension appear to protect against alcoholism. Much attention in genetic mapping experiments has focused on membrane-signaling proteins known to be affected by ethanol and on protein constituents of reward pathways in the brain. Polymorphisms associated with a relative insensitivity to alcohol and presumably thereby a greater risk of alcohol abuse have been identified in genes encoding an Îą subunit of the GABAA receptor, an M 2 muscarinic receptor, a serotonin transporter, adenylyl cyclase, and a potassium channel. The link between a polymorphism in an opioid receptor gene and a blunted response to naltrexone raises the possibility of genotype-guided pharmacotherapy for alcohol dependence.

MANAGEMENT OF ACUTE ALCOHOL INTOXICATION Nontolerant individuals who consume alcohol in large quantities develop typical effects of acute sedative-hypnotic drug overdose along with the cardiovascular effects previously described (vasodilation, tachycardia) and gastrointestinal irritation. Since tolerance is not


absolute, even individuals with chronic alcohol dependence may become severely intoxicated if sufficient alcohol is consumed. The most important goals in the treatment of acute alcohol intoxication are to prevent severe respiratory depression and aspiration of vomitus. Even with very high blood ethanol levels, survival is probable as long as the respiratory and cardiovascular systems can be supported. The average blood alcohol concentration in fatal cases is above 400 mg/dL; however, the lethal dose of alcohol varies because of varying degrees of tolerance. Electrolyte imbalances often need to be corrected and metabolic alterations may require treatment of hypoglycemia and ketoacidosis by administration of glucose. Thiamine is given to protect against Wernicke-Korsakoff syndrome. Patients who are dehydrated and vomiting should also receive electrolyte solutions. If vomiting is severe, large amounts of potassium may be required as long as renal function is normal.

MANAGEMENT OF ALCOHOL WITHDRAWAL SYNDROME Abrupt alcohol discontinuation in an individual with alcohol dependence leads to a characteristic syndrome of motor agitation, anxiety, insomnia, and reduction of seizure threshold. The severity of the syndrome is usually proportionate to the degree and duration of alcohol abuse. However, this can be greatly modified by the use of other sedatives as well as by associated factors (eg, diabetes, injury). In its mildest form, the alcohol withdrawal syndrome of increased pulse and blood pressure, tremor, anxiety, and insomnia occurs 6–8 hours after alcohol consumption is stopped (Figure 23–2). These effects usually lessen in 1–2 days, although some, such as anxiety and sleep disturbances, can be seen at decreasing levels for several months. In some patients, more severe acute reactions occur, with patients at risk of withdrawal seizures or alcoholic hallucinations during the first 1–5 days of withdrawal. Alcohol withdrawal is one of the most common causes of seizures in adults. Several days later, individuals can develop the syndrome of delirium tremens, which is characterized by delirium, agitation, autonomic nervous system instability, low-grade fever, and diaphoresis.

FIGURE 23–2 Time course of events during the alcohol withdrawal syndrome. The signs and symptoms that manifest earliest are anxiety, insomnia, tremor, palpitations, nausea, and anorexia as well as (in severe syndromes) hallucinations and seizures. Delirium tremens typically develops 48–72 hours after alcohol discontinuation. The earliest symptoms (anxiety, insomnia, etc) can persist, in a milder form, for several months after alcohol discontinuation. The major objective of drug therapy in the alcohol withdrawal period is prevention of seizures, delirium, and arrhythmias. Potassium, magnesium, and phosphate balance should be restored as rapidly as is consistent with renal function. Thiamine therapy is initiated in all cases. Individuals in mild alcohol withdrawal do not need any other pharmacologic assistance. Specific drug treatment for detoxification in more severe cases involves two basic principles: substituting a long-acting sedativehypnotic drug for alcohol and then gradually reducing (“tapering”) the dose of the long-acting drug. Because of their wide margin of


safety, benzodiazepines are preferred. The choice of a specific agent in this class is generally based on pharmacokinetic or economic considerations. Long-acting benzodiazepines, including chlordiazepoxide and diazepam, have the advantage of requiring less frequent dosing. Since their pharmacologically active metabolites are eliminated slowly, the long-acting drugs provide a built-in tapering effect. A disadvantage of the long-acting drugs is that they and their active metabolites may accumulate, especially in patients with compromised liver function. Short-acting drugs such as lorazepam and oxazepam are rapidly converted to inactive water-soluble metabolites that will not accumulate, and for this reason the short-acting drugs are especially useful in alcoholic patients with liver disease. Benzodiazepines can be administered orally in mild or moderate cases, or parenterally for patients with more severe withdrawal reactions. After the alcohol withdrawal syndrome has been treated acutely, sedative-hypnotic medications must be tapered slowly over several weeks. Complete detoxification is not achieved with just a few days of alcohol abstinence. Several months may be required for restoration of normal nervous system function, especially sleep.

TREATMENT OF ALCOHOLISM After detoxification, psychosocial therapy either in intensive inpatient or in outpatient rehabilitation programs serves as the primary treatment for alcohol dependence. Other psychiatric problems, most commonly depressive or anxiety disorders, often coexist with alcoholism and, if untreated, can contribute to the tendency of detoxified alcoholics to relapse. Treatment for these associated disorders with counseling and drugs can help decrease the rate of relapse for alcoholic patients. Three drugs—disulfiram, naltrexone, and acamprosate—have FDA approval for adjunctive treatment of alcohol dependence.

Naltrexone Naltrexone, a relatively long-acting opioid antagonist, blocks the effects at Îź-opioid receptors (see Chapter 31). Studies in experimental animals first suggested a link between alcohol consumption and opioids. Injection of small amounts of opioids was followed by an increase in alcohol drinking, whereas administration of opioid antagonists inhibited self-administration of alcohol. Naltrexone, both alone and in combination with behavioral counseling, has been shown in a number of short-term (12- to 16-week) placebo-controlled trials to reduce the rate of relapse to either drinking or alcohol dependence and to reduce craving for alcohol, especially in patients with high rates of naltrexone adherence. Naltrexone is approved by the FDA for treatment of alcohol dependence. Naltrexone is generally taken once a day in an oral dose of 50 mg for treatment of alcoholism. An extended-release formulation administered as an IM injection once every 4 weeks is also effective. The drug can cause dose-dependent hepatotoxicity and should be used with caution in patients with evidence of abnormalities in serum aminotransferase activity. The combination of naltrexone plus disulfiram should be avoided, since both drugs are potential hepatotoxins. Administration of naltrexone to patients who are physically dependent on opioids precipitates an acute withdrawal syndrome, so patients must be opioid-free before initiating naltrexone therapy. Naltrexone also blocks the therapeutic analgesic effects of usual doses of opioids.

Acamprosate Acamprosate has been used in Europe for a number of years to treat alcohol dependence and is approved for this use by the FDA. Like ethanol, acamprosate has many molecular effects including actions on GABA, glutamate, serotonergic, noradrenergic, and dopaminergic receptors. Probably its best-characterized actions are as a weak NMDA-receptor antagonist and a GABAA-receptor activator. In European clinical trials, acamprosate reduced short-term and long-term (more than 6 months) relapse rates when combined with psychotherapy. However, in a large American trial that compared acamprosate with naltrexone and with combined acamprosate and naltrexone therapy (the COMBINE study), acamprosate did not show a statistically significant effect alone or in combination with naltrexone. Acamprosate is administered as one or two enteric-coated 333 mg tablets three times daily. It is poorly absorbed, and food reduces its absorption even further. Acamprosate is widely distributed and is eliminated renally. It does not appear to participate in drug-drug interactions. The most common adverse effects are gastrointestinal (nausea, vomiting, diarrhea) and rash. It should not be used in patients with severe renal impairment.

Disulfiram Disulfiram causes extreme discomfort in patients who drink alcoholic beverages. Disulfiram alone has little effect; however, flushing, throbbing headache, nausea, vomiting, sweating, hypotension, and confusion occur within a few minutes after an individual taking disulfiram drinks alcohol. The effects may last 30 minutes in mild cases or several hours in severe ones. Disulfiram acts by inhibiting aldehyde dehydrogenase. Thus, alcohol is metabolized as usual, but acetaldehyde accumulates. Disulfiram is rapidly and completely absorbed from the gastrointestinal tract; however, a period of 12 hours is required for its full action. Its elimination rate is slow, so that its action may persist for several days after the last dose. The drug inhibits the metabolism of


many other therapeutic agents, including phenytoin, oral anticoagulants, and isoniazid. It should not be administered with medications that contain alcohol, including nonprescription medications such as those listed in Table 63–3. Disulfiram can cause small increases in hepatic transaminases. Its safety in pregnancy has not been demonstrated. Because adherence to disulfiram therapy is low and because the evidence from clinical trials for its effectiveness is weak, disulfiram is no longer commonly used.

Other Drugs Several other drugs have shown efficacy in maintaining abstinence and reducing craving in chronic alcoholism, although none has FDA approval yet for this use. Such drugs include ondansetron, a serotonin 5-HT3 -receptor antagonist (see Chapters 16, 62); topiramate, a drug used for partial and generalized tonic-clonic seizures (see Chapter 24); and baclofen, a GABA receptor antagonist used as a spasmolytic (see Chapter 27). Based on evidence from model systems, efforts are underway to explore agents that modulate cannabinoid CB1 receptors, corticotropin-releasing factor receptors, and GABA receptor systems, as well as several other possible targets. Rimonabant, a CB1 receptor antagonist, has been shown to suppress alcohol-related behaviors in animal models and is being tested in clinical trials of alcoholism.

PHARMACOLOGY OF OTHER ALCOHOLS Other alcohols related to ethanol have wide applications as industrial solvents and occasionally cause severe poisoning. Of these, methanol and ethylene glycol are two of the most common causes of intoxication. Isopropyl alcohol (isopropanol, rubbing alcohol) is another alcohol that is sometimes ingested when ethanol is not available. It produces coma and gastrointestinal irritation, nausea, and vomiting, but is not usually associated with retinal or renal injury.

METHANOL Methanol (methyl alcohol, wood alcohol) is widely used in the industrial production of synthetic organic compounds and as a constituent of many commercial solvents. In the home, methanol is most frequently found in the form of “canned heat” or in windshield-washing products. Poisonings occur from accidental ingestion of methanol-containing products or when it is misguidedly ingested as an ethanol substitute. Methanol can be absorbed through the skin or from the respiratory or gastrointestinal tract and is then distributed in body water. The primary mechanism of elimination of methanol in humans is by oxidation to formaldehyde, formic acid, and CO2 (Figure 23–3).


FIGURE 23–3 Methanol is converted to the toxic metabolites formaldehyde and formate by alcohol dehydrogenase and aldehyde dehydrogenase. By inhibiting alcohol dehydrogenase, fomepizole and ethanol reduce the formation of toxic metabolites. Animal species show great variability in mean lethal doses of methanol. The special susceptibility of humans to methanol toxicity is due to metabolism to formate and formaldehyde, not to methanol itself. Since the conversion of methanol to its toxic metabolites is relatively slow, there is often a delay of 6–30 hours before the appearance of severe toxicity. Physical findings in early methanol poisoning are generally nonspecific, such as inebriation and gastritis, and possibly an elevated osmolar gap (see Chapter 58). In severe cases, the odor of formaldehyde may be present on the breath or in the urine. After a delay, the most characteristic symptom in methanol poisoning—visual disturbance—occurs along with anion gap metabolic acidosis. The visual disturbance is frequently described as “like being in a snowstorm” and can progress to blindness. Changes in the retina may sometimes be detected on examination, but these are usually late. The development of bradycardia, prolonged coma, seizures, and resistant acidosis all imply a poor prognosis. The cause of death in fatal cases is sudden cessation of respiration. A serum methanol concentration higher than 20 mg/dL warrants treatment, and a concentration higher than 50 mg/dL is considered serious enough to require hemodialysis. Serum formate levels are a better indication of clinical pathology but are not widely available. The first treatment for methanol poisoning, as in all critical poisoning situations, is support of respiration. There are three specific modalities of treatment for severe methanol poisoning: suppression of metabolism by alcohol dehydrogenase to toxic products, hemodialysis to enhance removal of methanol and its toxic products, and alkalinization to counteract metabolic acidosis. The enzyme chiefly responsible for methanol oxidation in the liver is alcohol dehydrogenase (Figure 23–3). Fomepizole, an alcohol dehydrogenase inhibitor, is approved for the treatment of methanol and ethylene glycol poisoning. It is administered intravenously in a loading dose of 15 mg/kg followed by 10 mg/kg every 12 hours for 48 hours and then 15 mg/kg every 12 hours thereafter until the serum methanol level falls below 20–30 mg/dL. The dosage increase after 48 hours is based on evidence that fomepizole rapidly induces its own metabolism by the cytochrome P450 system. Patients undergoing hemodialysis are given fomepizole more frequently (6 hours after the loading dose and every 4 hours thereafter). Fomepizole appears to be safe during the short time it is administered for treatment of methanol or ethylene glycol poisoning. The most common adverse effects are burning at the infusion site, headache, nausea, and dizziness. Intravenous ethanol is an alternative to fomepizole. It has a higher affinity than methanol for alcohol dehydrogenase; thus, saturation of the enzyme with ethanol reduces formate production. Ethanol is used intravenously as treatment for methanol and ethylene glycol poisoning. The dose-dependent characteristics of ethanol metabolism and the variability of ethanol metabolism require frequent monitoring of blood ethanol levels to ensure appropriate alcohol concentration. In cases of severe poisoning, hemodialysis (discussed in Chapter 58) can be used to eliminate both methanol and formate from the blood. Two other measures are commonly taken. Because of profound metabolic acidosis in methanol poisoning, treatment with bicarbonate often is necessary. Since folate-dependent systems are responsible for the oxidation of formic acid to CO2 in humans (Figure 23–3), folinic and folic acid are often administered to patients poisoned with methanol, although this has never been fully tested in clinical studies.

ETHYLENE GLYCOL Polyhydric alcohols such as ethylene glycol (CH2 OHCH2 OH) are used as heat exchangers, in antifreeze formulations, and as industrial solvents. Young children and animals are sometimes attracted by the sweet taste of ethylene glycol and, rarely, it is ingested intentionally as an ethanol substitute or in attempted suicide. Although ethylene glycol itself is relatively harmless and eliminated by the kidney, it is metabolized to toxic aldehydes and oxalate. Three stages of ethylene glycol overdose occur. Within the first few hours after ingestion, there is transient excitation followed by CNS depression. After a delay of 4–12 hours, severe metabolic acidosis develops from accumulation of acid metabolites and lactate. Finally, deposition of oxalate crystals in renal tubules occurs, followed by delayed renal insufficiency. The key to the diagnosis of ethylene glycol poisoning is recognition of anion gap acidosis, osmolar gap, and oxalate crystals in the urine in a patient without visual symptoms. As with methanol poisoning, early fomepizole is the standard treatment for ethylene glycol poisoning. Intravenous treatment with fomepizole is initiated immediately, as described above for methanol poisoning, and continued until the patient’s serum ethylene glycol concentration drops below a toxic threshold (20–30 mg/dL). Intravenous ethanol is an alternative to fomepizole in ethylene glycol poisoning. Hemodialysis effectively removes ethylene glycol and its toxic metabolites and is recommended for patients with a serum ethylene glycol concentration above 50 mg/dL, significant metabolic acidosis, and significant renal impairment. Fomepizole has reduced the need for hemodialysis, especially in patients with less severe acidosis and intact renal function.

SUMMARY The Alcohols and Associated Drugs



PREPARATIONS AVAILABLE

REFERENCES Anton RF: Naltrexone for the management of alcohol dependence. N Engl J Med 2008;359:715. Anton RF et al: Combined pharmacotherapies and behavioral interventions for alcohol dependence: T he COMBINE study: A randomized controlled trial. JAMA 2006;295:2003. Brent J: Fomepizole for ethylene glycol and methanol poisoning. N Engl J Med 2009;360:2216. Brodie MS et al: Ethanol interactions with calcium-dependent potassium channels. Alcohol Clin Exp Res 2007;31:1625. CDC Fetal Alcohol Syndrome website: http://www.cdc.gov/ncbddd/fas/ Chen YC et al: Polymorphism of ethanol-metabolism genes and alcoholism: Correlation of allelic variations with the pharmacokinetic and pharmacodynamic consequences. Chem Biol Interact 2009;178:2. Colombo G et al: T he cannabinoid CB1 receptor antagonist, rimonabant, as a promising pharmacotherapy for alcohol dependence: Preclinical evidence. Mol Neurobiol 2007;36:102. Crabbe JC et al: Alcohol-related genes: Contributions from studies with genetically engineered mice. Addict Biol 2006;11:195. Das SK, Vasudevan DM: Alcohol-induced oxidative stress. Life Sci 2007;81:177. Edenberg HJ: T he genetics of alcohol metabolism: Role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health 2007;30:5. Heffernan T M: T he impact of excessive alcohol use on prospective memory: A brief review. Curr Drug Abuse Rev 2008;1:36. Heilig M, Egli M: Pharmacologic treatment of alcohol dependence: T arget symptoms and target mechanisms. Pharmacol T her 2006;111:855. Hendricson AW et al: Aberrant synaptic activation of N-methyl-D-aspartate receptors underlies ethanol withdrawal hyperexcitability. J Pharmacol Exp T her 2007;321:60. Johnson BA: Update on neuropharmacological treatments for alcoholism: Scientific basis and clinical findings. Biochem Pharmacol 2008;75:34.


Jonsson IM et al: Ethanol prevents development of destructive arthritis. Proc Natl Acad Sci USA 2007;104:258. Klatsky AL: Alcohol and cardiovascular diseases. Expert Rev Cardiovasc T her 2009;7:499. Lepik KJ et al: Adverse drug events associated with the antidotes for methanol and ethylene glycol poisoning: A comparison of ethanol and fomepizole. Ann Emerg Med 2009;53:439. Lobo IA, Harris RA: GABA(A) receptors and alcohol. Pharmacol Biochem Behav 2008;90:90. Mann K et al: Acamprosate: Recent findings and future research directions. Alcohol Clin Exp Res 2008;32:1105. Mayfield RD, Harris RA, Schuckit MA: Genetic factors influencing alcohol dependence. Br J Pharmacol 2008;154:275. National Institute on Alcohol Abuse and Alcoholism website: http://www.niaaa.nih.gov/ O’Keefe JH, Bybee KA, Lavie CJ: Alcohol and cardiovascular health: T he razor-sharp double-edged sword. J Am Coll Cardiol 2007:50:1009. Olson KR et al (editors): Poisoning and Drug Overdose, 6th ed. McGraw-Hill, 2012. Qiang M, Denny AD, T icku MK: Chronic intermittent ethanol treatment selectively alters N-methyl-D-aspartate receptor subunit surface expression in cultured cortical neurons. Mol Pharmacol 2007:72:95. Seitz HK, Stickel F: Molecular mechanisms of alcohol-mediated carcinogenesis. Nat Rev Cancer 2007;7:599. Shuckit MA: Alcohol-use disorders. Lancet 2009;373:492. Srisurapanont M, Jarusuraisin N: Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev 2005;(1):CD001867. T etrault JM, O’Connor PG: Substance abuse and withdrawal in the critical care setting. Crit Care Clin 2008;24:767. Wolf FW, Heberlein U: Invertebrate models of drug abuse. J Neurobiol 2003;54:161.

CASE STUDY ANSWER This young man exhibits classic signs and symptoms of acute alcohol poisoning, which is confirmed by the blood alcohol concentration. We do not know from the case whether the patient was tolerant to the effects of alcohol but note that his blood alcohol concentration was in the lethal range for a nontolerant individual. Death most likely resulted from respiratory and cardiovascular collapse prior to medical treatment, complicated by a chemical pneumonitis secondary to aspiration of vomitus. The treatment of acute alcohol poisoning includes standard supportive care of airway, breathing, and circulation (“ABCs,” see Chapter 58). Intravenous access would be obtained and used to administer dextrose and thiamine, as well as other electrolytes and vitamins. If a young, previously healthy individual receives medical care in time, supportive care will most likely be highly effective. As the patient recovers, it is important to be vigilant for signs and symptoms of the alcohol withdrawal syndrome.


CHAPTER

24 Antiseizure Drugs Roger J. Porter, MD, & Brian S. Meldrum, MB, PhD

CASE STUDY A 23-year-old woman presents to the office for consultation regarding her antiseizure medications. Seven years ago, this otherwise healthy young woman had a generalized tonic-clonic seizure (GTCS) at home. She was rushed to the emergency department, at which time she was alert but complained of headache. A consulting neurologist placed her on levetiracetam, 500 mg bid. Four days later, EEG showed rare right temporal sharp waves. MRI was normal. One year after this episode, a repeat EEG was unchanged, and levetiracetam was gradually increased to 1000 mg bid. The patient had no significant adverse effects from this dosage. At age 21, she had a second GTCS while in college; further discussion with her roommate at that time revealed a history of two recent episodes of 1–2 minutes of altered consciousness with lip smacking (complex partial seizures). A repeat EEG showed occasional right temporal spikes. What is one possible strategy for controlling her present symptoms?

Approximately 1% of the world’s population has epilepsy, the third most common neurologic disorder after dementia and stroke. Although standard therapy permits control of seizures in 80% of these patients, millions (500,000 people in the USA alone) have uncontrolled epilepsy. Epilepsy is a heterogeneous symptom complex—a chronic disorder characterized by recurrent seizures. Seizures are finite episodes of brain dysfunction resulting from abnormal discharge of cerebral neurons. The causes of seizures are many and include the full range of neurologic diseases—from infection to neoplasm and head injury. In some subgroups, heredity has proved to be a predominant factor. Single gene defects, usually of an autosomal dominant nature involving genes coding voltage-gated ion channels or GABAA receptors, have been shown to account for a small number of familial generalized epilepsies. Commonly, one family shows multiple epilepsy syndromes including, for example, febrile seizures, absence attacks, and juvenile myoclonic epilepsy. The antiseizure drugs described in this chapter are also used in patients with febrile seizures or with seizures occurring as part of an acute illness such as meningitis. The term “epilepsy” is not usually applied to such patients unless chronic seizures develop later. Seizures are occasionally caused by an acute underlying toxic or metabolic disorder, in which case appropriate therapy should be directed toward the specific abnormality, eg, hypocalcemia. In most cases of epilepsy, however, the choice of medication depends on the empiric seizure classification.

DRUG DEVELOPMENT FOR EPILEPSY For a long time it was assumed that a single antiepileptic drug (AED) could be developed for the treatment of all forms of epilepsy. However, the causes of epilepsy are extremely diverse, encompassing genetic and developmental defects and infective, traumatic, neoplastic, and degenerative disease processes. Drug therapy to date shows little evidence of etiologic specificity. There is some specificity according to seizure type (Table 24–1), which is most clearly seen with generalized seizures of the absence type. These are typically seen with 2–3 Hz spike-and-wave discharges on the electroencephalogram, which respond to ethosuximide and valproate but can be exacerbated by phenytoin and carbamazepine. Drugs acting selectively on absence seizures can be identified by animal screens, using either threshold pentylenetetrazol clonic seizures in mice or rats or mutant mice showing absence-like episodes (so-called lethargic, star-gazer, or tottering mutants). In contrast, the maximal electroshock (MES) test, with suppression of the tonic extensor phase, identifies drugs such as phenytoin, carbamazepine, and lamotrigine, which are active against generalized tonic-clonic seizures and complex partial seizures. The maximal electroshock test as the major initial screen for new drugs led predominantly to the early identification of drugs with a mechanism of action involving prolonged inactivation of the voltage-gated Na+ channel (see Chapter 14). Limbic seizures induced in rats by the process of electrical kindling (involving repeated episodes of focal electrical stimulation) probably provide a better screen for predicting efficacy in complex partial seizures.


TABLE 24–1 Classification of seizure types.

Existing antiseizure drugs provide adequate seizure control in about two thirds of patients. So-called “drug resistance” may be observed from the onset of attempted therapy or may develop after a period of relatively successful therapy. Explanations are being sought in terms of impaired access of the drugs to target sites or insensitivity of target molecules to them. In children, some severe seizure syndromes associated with progressive brain damage are very difficult to treat. In adults, some focal seizures are refractory to medications. Some, particularly in the temporal lobe, are amenable to surgical resection. Some of the drug-resistant population may respond to vagus nerve stimulation (VNS), a nonpharmacologic treatment for epilepsy now widely approved for treatment of patients with partial seizures. Another device approved in the USA for the treatment of medically refractory partial epilepsy is the responsive neurostimulator (RNS) system. The RNS neurostimulator is designed to detect abnormal electrical activity in the brain and deliver electrical brain stimulation to normalize activity before the patient experiences seizures. Other devices, using various paradigms of electrical stimulation, are in clinical development. One of these, a deep brain stimulation device, has been approved in Canada and in Europe, but not in the USA. New antiseizure drugs are being sought not only by the screening tests noted above but also by more focused approaches. Compounds are sought that act by one of three mechanisms: (1) enhancement of GABAergic (inhibitory) transmission, (2) diminution of excitatory (usually glutamatergic) transmission, or (3) modification of ionic conductances. Presynaptic effects on transmitter release appear particularly important, and some molecular targets are known, eg, SV2 A (see Figure 24–2). Although it is widely recognized that current antiseizure drugs are palliative rather than curative, successful strategies for identifying drugs that are either disease modifying or that prevent epileptogenesis have proved elusive. Neuronal targets for current and potential antiseizure drugs include both excitatory and inhibitory synapses. Figure 24–1 represents a glutamatergic (excitatory) synapse, and Figure 24–2 indicates targets in a GABAergic (inhibitory) synapse.



FIGURE 24–1 Molecular targets for antiseizure drugs at the excitatory, glutamatergic synapse. Presynaptic targets diminishing glutamate release include 1, voltage-gated (VG) Na+ channels (phenytoin, carbamazepine, lamotrigine, and lacosamide); 2, VG-Ca2+ channels (ethosuximide, lamotrigine, gabapentin, and pregabalin); 3, K+ channels (retigabine); synaptic vesicle proteins, 4, SV2 A (levetiracetam); and 5, CRMP-2, collapsin-response mediator protein-2. Postsynaptic targets include 6, AMPA receptors (blocked by phenobarbital, topiramate, lamotrigine, and perampanel) and 7, NMDA receptors (blocked by felbamate). EAAT, excitatory amino acid transporter; NTFs, neurotrophic factors; SV2 A, synaptic vesicular proteins. Red dots represent glutamate.



FIGURE 24–2 Molecular targets for antiseizure drugs at the inhibitory, GABAergic synapse. These include “specific” targets: 1, GABA transporters (especially GAT-1, tiagabine); 2, GABA-transaminase (GABA-T, vigabatrin); 3, GABAA receptors (benzodiazepines); potentially, 4, GABAB receptors; and 5, synaptic vesicular proteins (SV2 A). Effects may also be mediated by “nonspecific” targets such as by voltage-gated (VG) ion channels and synaptic proteins. IPSP, inhibitory postsynaptic potential. Blue dots represent GABA.

BASIC PHARMACOLOGY OF ANTISEIZURE DRUGS CHEMISTRY Until 1990, approximately 16 antiseizure drugs were available, and 13 of them can be classified into five very similar chemical groups: barbiturates, hydantoins, oxazolidinediones, succinimides, and acetylureas. These groups have in common a similar heterocyclic ring structure with a variety of substituents (Figure 24–3). For drugs with this basic structure, the substituents on the heterocyclic ring determine the pharmacologic class, either anti-MES or antipentylenetetrazol. Very small changes in structure can dramatically alter the mechanism of action and clinical properties of the compound. The remaining drugs in this older group—carbamazepine, valproic acid, and the benzodiazepines—are structurally dissimilar, as are the newer compounds marketed since 1990, ie, eslicarbazepine, felbamate, gabapentin, lacosamide, lamotrigine, levetiracetam, oxcarbazepine, perampanel, pregabalin, retigabine, rufinamide, stiripentol, tiagabine, topiramate, vigabatrin, and zonisamide.

FIGURE 24–3 Antiseizure heterocyclic ring structure. The X varies as follows: hydantoin derivatives, –N–; barbiturates, –C–N–; oxazolidinediones, –O–; succinimides, –C–; acetylureas, –NH2 (N connected to C2 ). R1 , R2 , and R3 vary within each subgroup.

PHARMACOKINETICS The antiseizure drugs exhibit many similar pharmacokinetic properties—even those whose structural and chemical properties are quite diverse—because most have been selected for oral activity and all must enter the central nervous system. Although many of these compounds are only slightly soluble, absorption is usually good, with 80–100% of the dose reaching the circulation. Most antiseizure drugs (other than phenytoin, tiagabine, and valproic acid) are not highly bound to plasma proteins. Antiseizure drugs are cleared chiefly by hepatic mechanisms, although they have low extraction ratios (see Chapter 3). Many are converted to active metabolites that are also cleared by the liver. These drugs are predominantly distributed into total body water. Plasma clearance is relatively slow; many antiseizure drugs are therefore considered to be medium to long acting. Some have half-lives longer than 12 hours. Many of the older antiseizure drugs are potent inducers of hepatic microsomal enzyme activity. Compliance is better with less frequent administration; thus extended-release formulations permitting once- or twice-daily administration may offer an advantage.

DRUGS USED IN PARTIAL SEIZURES & GENERALIZED TONIC-CLONIC SEIZURES The classic major drugs for partial and generalized tonic-clonic seizures are phenytoin (and congeners), carbamazepine, valproate, and the barbiturates. However, the availability of newer drugs—eslicarbazepine, lamotrigine, levetiracetam, gabapentin, oxcarbazepine, pregabalin, retigabine, topiramate, vigabatrin, lacosamide, and zonisamide—is altering clinical practice in countries where these compounds are available. The next section of the chapter is a description of major drugs from a historical and structural perspective. Factors involved in the clinical choice of drugs are described in the last section of the chapter.

PHENYTOIN Phenytoin is the oldest nonsedative antiseizure drug, introduced in 1938 after a systematic evaluation of compounds such as phenobarbital that altered electrically induced seizures in laboratory animals. It was known for decades as diphenylhydantoin.


Chemistry Phenytoin is a diphenyl-substituted hydantoin with the structure shown. It has much lower sedative properties than compounds with alkyl substituents at the 5 position. A more soluble prodrug of phenytoin, fosphenytoin, is available for parenteral use; this phosphate ester compound is rapidly converted to phenytoin in the plasma.

Mechanism of Action Phenytoin has major effects on several physiologic systems. It alters Na+, K+, and Ca2+ conductance, membrane potentials, and the concentrations of amino acids and the neurotransmitters norepinephrine, acetylcholine, and γ-aminobutyric acid (GABA). Studies with neurons in cell culture show that phenytoin blocks sustained high-frequency repetitive firing of action potentials (Figure 24–4). This effect is seen at therapeutically relevant concentrations. It is a use-dependent effect (see Chapter 14) on Na+ conductance, arising from preferential binding to—and prolongation of—the inactivated state of the Na+ channel. This effect is also seen with therapeutically relevant concentrations of carbamazepine, lamotrigine, and valproate and probably contributes to their antiseizure action in the electroshock model and in partial seizures. Phenytoin also blocks the persistent Na+ current, as do several other AEDs including valproate, topiramate, and ethosuximide.

FIGURE 24–4 Effects of three antiseizure drugs on sustained high-frequency firing of action potentials by cultured neurons. Intracellular recordings were made from neurons while depolarizing current pulses, approximately 0.75 s in duration, were applied (on-off step changes indicated by arrows). In the absence of drug, a series of high-frequency repetitive action potentials filled the entire duration of the current pulse. Phenytoin, carbamazepine, and sodium valproate all markedly reduced the number of action potentials elicited by the current pulses. (Adapted, with permission, from Macdonald RL, Meldrum BS: Principles of anti-epileptic drug action. In: Levy RH et al [editors]: Antiepileptic Drugs, 4th ed. Raven Press, 1995.)

In addition, phenytoin paradoxically causes excitation in some cerebral neurons. A reduction of calcium permeability, with inhibition of calcium influx across the cell membrane, may explain the ability of phenytoin to inhibit a variety of calcium-induced secretory processes, including release of hormones and neurotransmitters. Recording of excitatory and inhibitory postsynaptic potentials show that phenytoin decreases the synaptic release of glutamate and enhances the release of GABA. The mechanism of phenytoin’s action probably involves a combination of actions at several levels. At therapeutic concentrations, the major action of phenytoin is to block Na + channels and inhibit the generation of rapidly repetitive action potentials. Presynaptic actions on glutamate and GABA release probably arise from actions other than those on voltage-gated Na+ channels.


Clinical Uses Phenytoin is effective against partial seizures and generalized tonic-clonic seizures. In the latter, it appears to be effective against attacks that are either primary or secondary to another seizure type.

Pharmacokinetics Absorption of phenytoin is highly dependent on the formulation of the dosage form. Particle size and pharmaceutical additives affect both the rate and the extent of absorption. Absorption of phenytoin sodium from the gastrointestinal tract is nearly complete in most patients, although the time to peak may range from 3 to 12 hours. Absorption after intramuscular injection is unpredictable, and some drug precipitation in the muscle occurs; this route of administration is not recommended for phenytoin. In contrast, fosphenytoin, a more soluble phosphate prodrug of phenytoin, is well absorbed after intramuscular administration. Phenytoin is highly bound to plasma proteins. The total plasma level decreases when the percentage that is bound decreases, as in uremia or hypoalbuminemia, but correlation of free levels with clinical states remains uncertain. Drug concentration in cerebrospinal fluid is proportionate to the free plasma level. Phenytoin accumulates in brain, liver, muscle, and fat. Phenytoin is metabolized to inactive metabolites that are excreted in the urine. Only a very small proportion of the dose is excreted unchanged. The elimination of phenytoin is dose-dependent. At very low blood levels, phenytoin metabolism follows first-order kinetics. However, as blood levels rise within the therapeutic range, the maximum capacity of the liver to metabolize phenytoin is approached. Further increases in dosage, though relatively small, may produce very large changes in phenytoin concentrations (Figure 24–5). In such cases, the half-life of the drug increases markedly, steady state is not achieved in routine fashion (since the plasma level continues to rise), and patients quickly develop symptoms of toxicity.

FIGURE 24–5 Nonlinear relationship of phenytoin dosage and plasma concentrations. Five patients (identified by different symbols) received increasing dosages of phenytoin by mouth, and the steady-state serum concentration was measured at each dosage. The curves are not linear since, as the dosage increases, the metabolism is saturable. Note also the marked variation among patients in the serum levels achieved at any dosage. (Adapted, with permission, from Jusko WJ: Bioavailability and disposition kinetics of phenytoin in man. In: Kellaway P, Petersen I [editors]: Quantitative Analytic Studies in Epilepsy. Raven Press, 1977.)

The half-life of phenytoin varies from 12 to 36 hours, with an average of 24 hours for most patients in the low to mid therapeutic range. Much longer half-lives are observed at higher concentrations. At low blood levels, it takes 5–7 days to reach steady-state blood levels after every dosage change; at higher levels, it may be 4–6 weeks before blood levels are stable.

Therapeutic Levels & Dosage The therapeutic plasma level of phenytoin for most patients is between 10 and 20 mcg/mL. A loading dose can be given either orally or intravenously; the latter, using fosphenytoin, is the method of choice for convulsive status epilepticus (discussed later). When oral therapy is started, it is common to begin adults at a dosage of 300 mg/d, regardless of body weight. This may be acceptable in some patients, but


it frequently yields steady-state blood levels below 10 mcg/mL, which is the minimum therapeutic level for most patients. If seizures continue, higher doses are usually necessary to achieve plasma levels in the upper therapeutic range. Because of its dose-dependent kinetics, some toxicity may occur with only small increments in dosage. The phenytoin dosage should be increased each time by only 25– 30 mg in adults, and ample time should be allowed for the new steady state to be achieved before further increasing the dosage. A common clinical error is to increase the dosage directly from 300 mg/d to 400 mg/d; toxicity frequently occurs at a variable time thereafter. In children, a dosage of 5 mg/kg/d should be followed by readjustment after steady-state plasma levels are obtained. Two types of oral phenytoin sodium are currently available in the USA, differing in their respective rates of dissolution; one is absorbed rapidly and one more slowly. Only the slow-release extended-action formulation can be given in a single daily dosage, and care must be used when changing brands (see Preparations Available). Although a few patients being given phenytoin on a long-term basis have been proved to have low blood levels from poor absorption or rapid metabolism, the most common cause of low levels is poor compliance. Fosphenytoin sodium is available for intravenous or intramuscular use and replaces intravenous phenytoin sodium, a much less soluble form of the drug.

Drug Interactions & Interference with Laboratory Tests Drug interactions involving phenytoin are primarily related to protein binding or to metabolism. Since phenytoin is 90% bound to plasma proteins, other highly bound drugs, such as phenylbutazone and sulfonamides, can displace phenytoin from its binding site. In theory, such displacement may cause a transient increase in free drug. A decrease in protein binding—eg, from hypoalbuminemia—results in a decrease in the total plasma concentration of drug but not the free concentration. Intoxication may occur if efforts are made to maintain total drug levels in the therapeutic range by increasing the dose. The protein binding of phenytoin is decreased in the presence of renal disease. The drug has an affinity for thyroid-binding globulin, which confuses some tests of thyroid function; the most reliable screening test of thyroid function in patients taking phenytoin appears to be measurement of thyroid-stimulating hormone (TSH). Phenytoin has been shown to induce microsomal enzymes responsible for the metabolism of a number of drugs. Autostimulation of its own metabolism, however, appears to be insignificant.

Toxicity Dose-related adverse effects caused by phenytoin are often similar to those caused by other antiseizure drugs in this group, making differentiation difficult in patients receiving multiple drugs. Nystagmus occurs early, as does loss of smooth extraocular pursuit movements, but neither is an indication for decreasing the dose. Diplopia and ataxia are the most common dose-related adverse effects requiring dosage adjustment; sedation usually occurs only at considerably higher levels. Gingival hyperplasia and hirsutism occur to some degree in most patients; the latter can be especially unpleasant in women. Long-term use is associated in some patients with coarsening of facial features and with mild peripheral neuropathy, usually manifested by diminished deep tendon reflexes in the lower extremities. Long-term use may also result in abnormalities of vitamin D metabolism, leading to osteomalacia. Low folate levels and megaloblastic anemia have been reported, but the clinical importance of these observations is unknown. Idiosyncratic reactions to phenytoin are relatively rare. A skin rash may indicate hypersensitivity of the patient to the drug. Fever may also occur, and in rare cases the skin lesions may be severe and exfoliative. Lymphadenopathy may be difficult to distinguish from malignant lymphoma, and although some studies suggest a causal relationship between phenytoin and Hodgkin’s disease, the data are far from conclusive. Hematologic complications are exceedingly rare, although agranulocytosis has been reported in combination with fever and rash.

MEPHENYTOIN, ETHOTOIN, & PHENACEMIDE Many congeners of phenytoin have been synthesized, but only three have been marketed in the USA, and one of these (phenacemide) has been withdrawn. The other two congeners, mephenytoin and ethotoin, like phenytoin, appear to be most effective against generalized tonic-clonic seizures and partial seizures. No well-controlled clinical trials have documented their effectiveness. The incidence of severe reactions such as dermatitis, agranulocytosis, or hepatitis is higher for mephenytoin than for phenytoin. Ethotoin may be recommended for patients who are hypersensitive to phenytoin, but larger doses are required. The adverse effects and toxicity are generally less severe than those associated with phenytoin, but the drug appears to be less effective. Both ethotoin and mephenytoin share with phenytoin the property of saturable metabolism within the therapeutic dosage range. Careful monitoring of the patient during dosage alterations with either drug is essential. Mephenytoin is metabolized to 5,5ethylphenylhydantoin via demethylation. This metabolite, nirvanol, contributes most of the antiseizure activity of mephenytoin. Both mephenytoin and nirvanol are hydroxylated and undergo subsequent conjugation and excretion. Therapeutic levels for mephenytoin range from 5 to 16 mcg/mL, and levels above 20 mcg/mL are considered toxic. Therapeutic blood levels of nirvanol are between 25 and 40 mcg/mL. A therapeutic range for ethotoin has not been established.


CARBAMAZEPINE Closely related to imipramine and other antidepressants, carbamazepine is a tricyclic compound effective in treatment of bipolar depression. It was initially marketed for the treatment of trigeminal neuralgia but has proved useful for epilepsy as well.

Chemistry Although not obvious from a two-dimensional representation of its structure, carbamazepine has many similarities to phenytoin. The ureide moiety (–N–CO–NH2 ) in the heterocyclic ring of most antiseizure drugs is also present in carbamazepine. Three-dimensional structural studies indicate that its spatial conformation is similar to that of phenytoin.

Mechanism of Action The mechanism of action of carbamazepine appears to be similar to that of phenytoin. Like phenytoin, carbamazepine shows activity against maximal electroshock seizures. Carbamazepine, like phenytoin, blocks Na+ channels at therapeutic concentrations and inhibits high-frequency repetitive firing in neurons in culture (Figure 24–4) . It also acts presynaptically to decrease synaptic transmission. Potentiation of a voltage-gated K+ current has also been described. These effects probably account for the anticonvulsant action of carbamazepine. Binding studies show that carbamazepine interacts with adenosine receptors, but the functional significance of this observation is not known.

Clinical Uses Although carbamazepine has long been considered a drug of choice for both partial seizures and generalized tonic-clonic seizures, some of the newer antiseizure drugs are beginning to displace it from this role. Carbamazepine is not sedative in its usual therapeutic range. The drug is also very effective in some patients with trigeminal neuralgia, although older patients may tolerate higher doses poorly, with ataxia and unsteadiness. Carbamazepine is also useful for controlling mania in some patients with bipolar disorder.

Pharmacokinetics The rate of absorption of carbamazepine varies widely among patients, although almost complete absorption apparently occurs in all. Peak levels are usually achieved 6–8 hours after administration. Slowing absorption by giving the drug after meals helps the patient tolerate larger total daily doses. Distribution is slow, and the volume of distribution is roughly 1 L/kg. The drug is approximately 70% bound to plasma proteins; no displacement of other drugs from protein binding sites has been observed. Carbamazepine has a very low systemic clearance of approximately 1 L/kg/d at the start of therapy. The drug has a notable ability to induce microsomal enzymes. Typically, the half-life of 36 hours observed in subjects after an initial single dose decreases to as little as 8– 12 hours in subjects receiving continuous therapy. Considerable dosage adjustments are thus to be expected during the first weeks of therapy. Carbamazepine also alters the clearance of other drugs (see below). Carbamazepine is completely metabolized in humans to several derivatives. One of these, carbamazepine-10,11-epoxide, has been shown to have anticonvulsant activity. The contribution of this and other metabolites to the clinical activity of carbamazepine is unknown.

Therapeutic Levels & Dosage Carbamazepine is available only in oral form. The drug is effective in children, in whom a dosage of 15–25 mg/kg/d is appropriate. In adults, daily doses of 1 g or even 2 g are tolerated. Higher dosage is achieved by giving multiple divided doses daily. Extended-release preparations permit twice-daily dosing for most patients. In patients in whom the blood is drawn just before the morning dose (trough


level), the therapeutic level is usually 4–8 mcg/mL. Although many patients complain of diplopia at drug levels above 7 mcg/mL, others can tolerate levels above 10 mcg/mL, especially with monotherapy. Extended-release formulations that overcome some of these issues are now available.

Drug Interactions Drug interactions involving carbamazepine are almost exclusively related to the drug’s enzyme-inducing properties. As noted previously, the increased metabolic capacity of the hepatic enzymes may cause a reduction in steady-state carbamazepine concentrations and an increased rate of metabolism of other drugs, eg, primidone, phenytoin, ethosuximide, valproic acid, and clonazepam. Other drugs such as valproic acid may inhibit carbamazepine clearance and increase steady-state carbamazepine blood levels. Other anticonvulsants, however, such as phenytoin and phenobarbital, may decrease steady-state concentrations of carbamazepine through enzyme induction. No clinically significant protein-binding interactions have been reported.

Toxicity The most common dose-related adverse effects of carbamazepine are diplopia and ataxia. The diplopia often occurs first and may last less than an hour during a particular time of day. Rearrangement of the divided daily dose can often remedy this complaint. Other doserelated complaints include mild gastrointestinal upsets, unsteadiness, and, at much higher doses, drowsiness. Hyponatremia and water intoxication have occasionally occurred and may be dose related. Considerable concern exists regarding the occurrence of idiosyncratic blood dyscrasias with carbamazepine, including fatal cases of aplastic anemia and agranulocytosis. Most of these have been in elderly patients with trigeminal neuralgia, and most have occurred within the first 4 months of treatment. The mild and persistent leukopenia seen in some patients is not necessarily an indication to stop treatment but requires careful monitoring. The most common idiosyncratic reaction is an erythematous skin rash; other responses such as hepatic dysfunction are unusual.

OXCARBAZEPINE Oxcarbazepine is closely related to carbamazepine and is useful in the same seizure types, but it may have an improved toxicity profile. Oxcarbazepine has a half-life of only 1–2 hours. Its activity, therefore, resides almost exclusively in the 10-hydroxy metabolite (especially the S(+) enantiomer, eslicarbazepine), to which it is rapidly converted and which has a half-life similar to that of carbamazepine, ie, 8–12 hours. The drug is mostly excreted as the glucuronide of the 10-hydroxy metabolite.

Oxcarbazepine is less potent than carbamazepine, both in animal models of epilepsy and in epileptic patients; clinical doses of oxcarbazepine may need to be 50% higher than those of carbamazepine to obtain equivalent seizure control. Some studies report fewer hypersensitivity reactions to oxcarbazepine, and cross-reactivity with carbamazepine does not always occur. Furthermore, the drug appears to induce hepatic enzymes to a lesser extent than carbamazepine, minimizing drug interactions. Although hyponatremia may occur more commonly with oxcarbazepine than with carbamazepine, most adverse effects that occur with oxcarbazepine are similar in character to reactions reported with carbamazepine.

ESLICARBAZINE Eslicarbazepine acetate (ESL) is a prodrug that has been approved as adjunctive therapy in adults with partial-onset seizures, with or without secondary generalization. ESL is more rapidly converted to S(+)-licarbazine (eslicarbazine) than is oxcarbazepine; clearly both prodrugs have the same metabolite as active product. The mechanism of action of carbamazepine, oxcarbazepine, and ESL appears to


be the same, ie, blocking of voltage-gated Na+ channels. The R(−) enantiomer has some activity, but much less than its counterpart. Clinically, the drug is similar to carbamazepine and oxcarbazepine in its spectrum of action, but it is less well studied in other possible indications. A possible advantage of ESL is its once-daily dosing regimen. The measured half-life of the S(+) enantiomer is 9–11 hours. The drug is administered at a dosage of 400–1200 mg/d; titration is typically required for the higher doses. Minimal drug level effects are observed with co-administration of carbamazepine, levetiracetam, lamotrigine, topiramate, and valproate. Oral contraceptives may be less effective with concomitant ESL administration.

PHENOBARBITAL Aside from the bromides, phenobarbital is the oldest of the currently available antiseizure drugs. Although it has long been considered one of the safest of the antiseizure agents, the use of other medications with lesser sedative effects has been urged. Many consider the barbiturates the drugs of choice for seizures only in infants.

Chemistry The four derivatives of barbituric acid clinically useful as antiseizure drugs are phenobarbital, mephobarbital, metharbital, and primidone. The first three are so similar that they are considered together. Metharbital is methylated barbital, and mephobarbital is methylated phenobarbital; both are demethylated in vivo. The pKas of these three weak acid compounds range from 7.3 to 7.9. Slight changes in the normal acid-base balance, therefore, can cause significant fluctuation in the ratio of the ionized to the nonionized species. This is particularly important for phenobarbital, the most commonly used barbiturate, whose pKa is similar to the plasma pH of 7.4. The three-dimensional conformations of the phenobarbital and N-methylphenobarbital molecules are similar to that of phenytoin. Both compounds possess a phenyl ring and are active against partial seizures.

Mechanism of Action The exact mechanism of action of phenobarbital is unknown, but enhancement of inhibitory processes and diminution of excitatory transmission probably contribute significantly. Recent data indicate that phenobarbital may selectively suppress abnormal neurons. Like phenytoin, phenobarbital suppresses high-frequency repetitive firing in neurons in culture through an action on Na+ conductance, but only at high concentrations. Also at high concentrations, barbiturates block some Ca 2+ currents (L-type and N-type). Phenobarbital binds to an allosteric regulatory site on the GABAA receptor, and it enhances the GABA receptor-mediated current by prolonging the openings of the Cl– channels (see Chapter 22). Phenobarbital can also decrease excitatory responses. An effect on glutamate release is probably more significant than blockade of AMPA responses (see Chapter 21). Both the enhancement of GABA-mediated inhibition and the reduction of glutamate-mediated excitation are seen with therapeutically relevant concentrations of phenobarbital.

Clinical Uses Phenobarbital is useful in the treatment of partial seizures and generalized tonic-clonic seizures, although the drug is often tried for virtually every seizure type, especially when attacks are difficult to control. There is little evidence for its effectiveness in generalized seizures such as absence, atonic attacks, and infantile spasms; it may worsen certain patients with these seizure types. Some physicians prefer either metharbital (no longer readily available) or mephobarbital (especially the latter) to phenobarbital because of supposed decreased adverse effects. Only anecdotal data are available to support such comparisons.

Pharmacokinetics, Therapeutic Levels, & Dosage For pharmacokinetics, drug interactions, and toxicity of phenobarbital, see Chapter 22. The therapeutic levels of phenobarbital in most patients range from 10 mcg/mL to 40 mcg/mL. Documentation of effectiveness is best in febrile seizures, and levels below 15 mcg/mL appear ineffective for prevention of febrile seizure recurrence. The upper end of the therapeutic range is more difficult to define because many patients appear to tolerate chronic levels above 40 mcg/mL.

PRIMIDONE Primidone, or 2-desoxyphenobarbital (Figure 24–6), was first marketed in the early 1950s. It was later reported that primidone was metabolized to phenobarbital and phenylethylmalonamide (PEMA). All three compounds are active anticonvulsants.


FIGURE 24–6 Primidone and its active metabolites.

Mechanism of Action Although primidone is converted to phenobarbital, the mechanism of action of primidone itself may be more like that of phenytoin.

Clinical Uses Primidone, like its metabolites, is effective against partial seizures and generalized tonic-clonic seizures and may be more effective than phenobarbital. It was previously considered to be the drug of choice for complex partial seizures, but later studies of partial seizures in adults strongly suggest that carbamazepine and phenytoin are superior to primidone. Attempts to determine the relative potencies of the parent drug and its two metabolites have been conducted in newborn infants, in whom drug-metabolizing enzyme systems are very immature and in whom primidone is only slowly metabolized. Primidone has been shown to be effective in controlling seizures in this group and in older patients beginning treatment with primidone; older patients show seizure control before phenobarbital concentrations reach the therapeutic range. Finally, studies of maximal electroshock seizures in animals suggest that primidone has an anticonvulsant action independent of its conversion to phenobarbital and PEMA (the latter is relatively weak).

Pharmacokinetics Primidone is completely absorbed, usually reaching peak concentrations about 3 hours after oral administration, although considerable variation has been reported. Primidone is generally distributed in total body water, with a volume of distribution of 0.6 L/kg. It is not highly bound to plasma proteins; approximately 70% circulates as unbound drug. Primidone is metabolized by oxidation to phenobarbital, which accumulates very slowly, and by scission of the heterocyclic ring to form PEMA (Figure 24–6). Both primidone and phenobarbital also undergo subsequent conjugation and excretion. Primidone has a larger clearance than most other antiseizure drugs (2 L/kg/d), corresponding to a half-life of 6–8 hours. PEMA clearance is approximately half that of primidone, but phenobarbital has a very low clearance (see Table 3–1). The appearance of phenobarbital corresponds to the disappearance of primidone. Phenobarbital therefore accumulates very slowly but eventually reaches therapeutic concentrations in most patients when therapeutic doses of primidone are administered. During chronic therapy, phenobarbital levels derived from primidone are usually two to three times higher than primidone levels.

Therapeutic Levels & Dosage


Primidone is most efficacious when plasma levels are in the range of 8–12 mcg/mL. Concomitant levels of its metabolite, phenobarbital, at steady state usually vary from 15 to 30 mcg/mL. Dosages of 10–20 mg/kg/d are necessary to obtain these levels. It is very important, however, to start primidone at low doses and gradually increase over days to a few weeks to avoid prominent sedation and gastrointestinal complaints. When adjusting doses of the drug, it is important to remember that the parent drug reaches steady state rapidly (30–40 hours), but the active metabolites phenobarbital (20 days) and PEMA (3–4 days) reach steady state much more slowly.

Toxicity The dose-related adverse effects of primidone are similar to those of its metabolite, phenobarbital, except that drowsiness occurs early in treatment and may be prominent if the initial dose is too large. Gradual increments are indicated when starting the drug in either children or adults.

FELBAMATE Felbamate has been approved and marketed in the USA and in some European countries. Although it is effective in some patients with partial seizures, the drug causes aplastic anemia and severe hepatitis at unexpectedly high rates and has been relegated to the status of a third-line drug for refractory cases. Felbamate appears to have multiple mechanisms of action. It produces a use-dependent block of the NMDA receptor, with selectivity for the NR1-2B subtype. It also produces a barbiturate-like potentiation of GABAA receptor responses. Felbamate has a halflife of 20 hours (somewhat shorter when administered with either phenytoin or carbamazepine) and is metabolized by hydroxylation and conjugation; a significant percentage of the drug is excreted unchanged in the urine. When added to treatment with other antiseizure drugs, felbamate increases plasma phenytoin and valproic acid levels but decreases levels of carbamazepine.

In spite of the seriousness of the adverse effects, thousands of patients worldwide utilize this medication. Usual dosages are 2000– 4000 mg/d in adults, and effective plasma levels range from 30 mcg/mL to 100 mcg/mL. In addition to its usefulness in partial seizures, felbamate has proved effective against the seizures that occur in Lennox-Gastaut syndrome.

GABAPENTIN & PREGABALIN Gabapentin is an amino acid, an analog of GABA, that is effective against partial seizures. Originally planned as a spasmolytic, it was found to be more effective as an antiseizure drug. Pregabalin is another GABA analog, closely related to gabapentin, and has been approved for both antiseizure activity and for its analgesic properties.


Mechanism of Action In spite of their close structural resemblance to GABA, gabapentin and pregabalin do not act directly on GABA receptors. They may, however, modify the synaptic or nonsynaptic release of GABA. An increase in brain GABA concentration is observed in patients receiving gabapentin. Gabapentin is transported into the brain by the L-amino acid transporter. Gabapentin and pregabalin bind avidly to the α2δ subunit of voltage-gated N-type Ca2+ channels. This appears to underlie the main mechanism of action, which is decreasing Ca2+ entry, with a predominant effect on presynaptic channels. A decrease in the synaptic release of glutamate provides the antiepileptic effect.

Clinical Uses Gabapentin is effective as an adjunct against partial seizures and generalized tonic-clonic seizures at dosages that range up to 2400 mg/d in controlled clinical trials. Open follow-up studies permitted dosages up to 4800 mg/d, but data are inconclusive on the effectiveness or tolerability of such doses. Monotherapy studies also document some efficacy. Some clinicians have found that very high dosages are needed to achieve improvement in seizure control. Effectiveness in other seizure types has not been well demonstrated. Gabapentin has also been promoted for the treatment of neuropathic pain and is now indicated for postherpetic neuralgia in adults at doses of 1800 mg and above. The most common adverse effects are somnolence, dizziness, ataxia, headache, and tremor. Pregabalin is approved for the adjunctive treatment of partial seizures, with or without secondary generalization; controlled clinical trials have documented its effectiveness. It is available only in oral form, and the dosage ranges from 150 to 600 mg/d, usually in two or three divided doses. Pregabalin is also approved for use in neuropathic pain, including painful diabetic peripheral neuropathy and postherpetic neuralgia. It is the first drug in the USA approved for fibromyalgia. In Europe it is also approved for generalized anxiety disorder.

Pharmacokinetics Gabapentin is not metabolized and does not induce hepatic enzymes. Absorption is nonlinear and dose-dependent at very high doses, but the elimination kinetics are linear. The drug is not bound to plasma proteins. Drug-drug interactions are negligible. Elimination is via renal mechanisms; the drug is excreted unchanged. The half-life is relatively short, ranging from 5 to 8 hours; the drug is typically administered two or three times per day. Pregabalin, like gabapentin, is not metabolized and is almost entirely excreted unchanged in the urine. It is not bound to plasma proteins and has virtually no drug-drug interactions, again resembling the characteristics of gabapentin. Likewise, other drugs do not affect the pharmacokinetics of pregabalin. The half-life of pregabalin ranges from about 4.5 hours to 7.0 hours, thus requiring more than once-daily dosing in most patients.

LACOSAMIDE Lacosamide is an amino acid-related compound that has been studied in both pain syndromes and partial seizures. The drug was approved in Europe and the USA in 2008 for the treatment of partial seizures.

Mechanism of Action Activity resides in the R(−) enantiomer. It does not act directly on GABA or glutamate receptors. Lacosamide enhances slow inactivation of voltage-gated Na+ channels (in contrast to the prolongation of fast inactivation shown by other AEDs). Slow inactivation (with a half-time of around 100 msec) does not result in complete blockade of Na+ channels. Nevertheless, the antiseizure effects (and the CNS side effects) of lacosamide are additive to those of established AEDs acting by prolonging inactivation of the Na + channel. Although lacosamide was previously thought to bind to the collapsin-response mediator protein, CRMP-2, thereby blocking the effect of neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) on axonal and dendritic growth, recent evidence suggests that such binding does not occur.

Clinical Uses Lacosamide is approved as adjunctive therapy in the treatment of partial-onset seizures with or without secondary generalization in patients with epilepsy who are age 16–17 years and older. Clinical trials include three multicenter, randomized placebo-controlled studies with more than 1300 patients. Treatment was effective at both 200 and 400 mg/d. Adverse effects were dizziness, headache, nausea, and diplopia. In the open-label follow-up study, at dosages ranging from 100 to 800 mg/d, many patients continued lacosamide treatment for 24–30 months. The drug is typically administered twice daily, beginning with 50 mg doses and increasing by 100 mg increments


weekly. An intravenous formulation provides short-term replacement for the oral drug. The oral solution is a source of phenylalanine.

Pharmacokinetics Oral lacosamide is rapidly and completely absorbed in adults, with no food effect. Bioavailability is nearly 100%. The plasma concentrations are proportional to dosage up to 800 mg orally. Peak concentrations occur from 1 to 4 hours after oral dosing, with an elimination half-life of 13 hours. There are no active metabolites and protein binding is minimal. Lacosamide does not induce or inhibit cytochrome P450 isoenzymes, so drug interactions are negligible.

LAMOTRIGINE Lamotrigine was developed when some investigators thought that the antifolate effects of certain antiseizure drugs (eg, phenytoin) might contribute to their effectiveness. Several phenyltriazines were developed, and though their antifolate properties were weak, some were active in seizure screening tests.

Mechanism of Action Lamotrigine, like phenytoin, suppresses sustained rapid firing of neurons and produces a voltage- and use-dependent blockade of Na+ channels. This action probably explains lamotrigine’s efficacy in focal epilepsy. It appears likely that lamotrigine also inhibits voltagegated Ca2+ channels, particularly the N- and P/Q-type channels, which would account for its efficacy in primary generalized seizures in childhood, including absence attacks. Lamotrigine also decreases the synaptic release of glutamate.

Clinical Uses Although most controlled studies have evaluated lamotrigine as add-on therapy, it is generally agreed that the drug is effective as monotherapy for partial seizures, and lamotrigine is now widely prescribed for this indication. The drug is also active against absence and myoclonic seizures in children and is approved for seizure control in the Lennox-Gastaut syndrome. Lamotrigine is also effective for bipolar disorder. Adverse effects include dizziness, headache, diplopia, nausea, somnolence, and skin rash. The rash is considered a typical hypersensitivity reaction. Although the risk of rash may be diminished by introducing the drug slowly, pediatric patients are at greatest risk, some studies suggest that a potentially life-threatening dermatitis will develop in 1–2% of pediatric patients.

Pharmacokinetics Lamotrigine is almost completely absorbed and has a volume of distribution in the range of 1–1.4 L/kg. Protein binding is only about 55%. The drug has linear kinetics and is metabolized primarily by glucuronidation to the 2-N-glucuronide, which is excreted in the urine. Lamotrigine has a half-life of approximately 24 hours in normal volunteers; this decreases to 13–15 hours in patients taking enzymeinducing drugs. Lamotrigine is effective against partial seizures in adults at dosages typically between 100 and 300 mg/d and with a therapeutic blood level near 3 mcg/mL. Valproate causes a twofold increase in the drug’s half-life; in patients receiving valproate, the initial dosage of lamotrigine must be reduced to 25 mg every other day.

LEVETIRACETAM


Levetiracetam is a piracetam analog that is ineffective against seizures induced by maximum electroshock or pentylenetetrazol but has prominent activity in the kindling model. This is the first major drug with this unusual preclinical profile that is effective against partial seizures. Brivaracetam, an analog of levetiracetam, is in clinical trials.

Mechanism of Action Levetiracetam binds selectively to the synaptic vesicular protein SV2 A. The function of this protein is not understood but it is likely that levetiracetam modifies the synaptic release of glutamate and GABA through an action on vesicular function. In addition, levetiracetam inhibits N-type calcium channels and inhibits calcium release from intracellular stores.

Clinical Uses Levetiracetam is marketed for the adjunctive treatment of partial seizures in adults and children for primary generalized tonic-clonic seizures and for the myoclonic seizures of juvenile myoclonic epilepsy. Adult dosing can begin with 500 or 1000 mg/d. The dosage can be increased every 2–4 weeks by 1000 mg to a maximum dosage of 3000 mg/d. The drug is dosed twice daily. Adverse effects include somnolence, asthenia, ataxia, and dizziness. Less common but more serious are mood and behavioral changes; psychotic reactions are rare. Drug interactions are minimal; levetiracetam is not metabolized by cytochrome P450. Oral formulations include extended-release tablets; an intravenous preparation is also available.

Pharmacokinetics Oral absorption of levetiracetam is nearly complete; it is rapid and unaffected by food, with peak plasma concentrations in 1.3 hours. Kinetics are linear. Protein binding is less than 10%. The plasma half-life is 6–8 hours, but may be longer in the elderly. Two thirds of the drug is excreted unchanged in the urine; the drug has no known active metabolites.

PERAMPANEL Perampanel is an orally active AMPA antagonist approved for the treatment of partial seizures.

Mechanism of Action Perampanel acts selectively at postsynaptic AMPA receptors ( Figure 24–1). It binds to an allosteric site on the glutamate-gated Na+/K+ AMPA channel and is therefore noncompetitive in its action. Whereas blockade of the NMDA receptor shortens the duration of repetitive discharge in model neuronal systems, blockade of AMPA receptors appears to prevent such discharge.

Clinical Uses Perampanel is approved for the adjunctive treatment of partial seizures with or without secondary generalization in patients 12 years of age or older. Three phase 3 studies, including 1480 patients, confirmed the effectiveness of the drug using once-daily administration. Effective doses ranged from 4 to 12 mg/d. Although the drug was generally well tolerated, a small number of patients experienced serious or life-threating behavioral adverse reactions including aggression, hostility, irritability, and anger, with or without a previous history of psychiatric disorders. More common adverse effects were dizziness, somnolence, and headache. Falls were more common at higher doses. Although a rash occurred in 1–2% of patients, all had benign outcomes when the drug was discontinued.

Pharmacokinetics


Perampanel has a long half-life, typically ranging from 70 to 110 hours, which permits once-daily dosing. Steady state is not achieved for 2–3 weeks, which has substantial implications for dosage changes. The kinetics are linear in the dose range of 2–12 mg/d. The half-life is prolonged in moderate hepatic failure. Absorption is rapid and the drug is fully bioavailable. Although food slows the rate of absorption, the extent (AUC) is not affected. Perampanel is 95% bound to plasma proteins. The drug is extensively metabolized via initial oxidation and subsequent glucuronidation. Although oxidative metabolism appears to be primarily mediated by CYP3A4 and CYP3A5, these may not be the only pathways involved.

Drug Interactions The most significant drug interactions with perampanel are with potent CYP3A inducer antiseizure drugs such as carbamazepine, oxcarbazepine, and phenytoin. Interactions are also significant with alcohol and with oral contraceptives containing levonorgestrel. Potent CYP3A inducers may increase the clearance of perampanel by 50–70%, requiring careful consideration when using these drugs concomitantly. When perampanel was administered with carbamazepine, the half-life decreased from 105 hours to 25 hours. Of somewhat lesser concern is the potential for strong P450 inhibitors to increase the levels of perampanel.

RETIGABINE (EZOGABINE) Retigabine (ezogabine in the USA) is approved for the adjunctive treatment of partial-onset seizures in adults. It is a potassium-channel facilitator and unique in its mechanism of action. Absorption is not affected by food and kinetics are linear; drug interactions are minimal. Doses range from 600 to 1200 mg/d, with 900 mg/day expected to be the median. The current dosage form requires three-times-per-day administration, and the dose must be titrated in most patients. Most adverse effects are dose-related and include dizziness, somnolence, blurred vision, confusion, and dysarthria. Bladder dysfunction, mostly mild and related to the drug’s mechanism of action, was observed in 8-9% of patients in the clinical trials. In 2013, reports began to appear of blue pigmentation, primarily on the skin and lips; the problem is rather common, occurring in about one third of patients on long-term therapy. Retinal pigment abnormalities are less common but may occur independent of skin changes. Decreased visual acuity has been reported, but documentation is lacking. Nevertheless, any of the above symptoms are reasons to consider the discontinuation of retigabine. Regulatory agencies have recommended use of retigabine only in cases where other antiseizure drugs are not adequate or not tolerated. The FDA recently announced changes in the labeling of ezogabine to warn about the risks of retinal abnormalities, possible vision loss, and bluish skin discoloration, all of which could be permanent. More information is available at http://secure.medicalletter.org/w1430d#sthash.BN17EI1Y.dpuf.

RUFINAMIDE Rufinamide is a triazole derivative with little similarity to other antiseizure drugs.

Mechanism of Action Rufinamide is protective in the maximal electroshock and pentylenetetrazol tests in rats and mice. It decreases sustained high-frequency firing of neurons in vitro and is thought to prolong the inactive state of the Na+ channel. Significant interactions with GABA systems or metabotropic glutamate receptors have not been seen.

Clinical Uses Rufinamide is approved in the USA for adjunctive treatment of seizures associated with the Lennox-Gastaut syndrome in patients age 4 years and older. The drug is effective against all seizure types in this syndrome, especially against tonic-atonic seizures. Recent data also suggest it may be effective against partial seizures. Treatment in children is typically started at 10 mg/kg/d in two equally divided doses and gradually increased to 45 mg/kg/d or 3200 mg/d, whichever is lower. Adults can begin with 400–800 mg/d in two equally divided doses up to a maximum of 3200 mg/d as tolerated. The drug should be given with food. The most common adverse events are


somnolence, vomiting, pyrexia, and diarrhea.

Pharmacokinetics Rufinamide is well absorbed, but plasma concentrations peak between 4 and 6 hours. The half-life is 6–10 hours, and minimal plasma protein binding is observed. Although cytochrome P450 enzymes are not involved, the drug is extensively metabolized to inactive products. Most of the drug is excreted in the urine; an acid metabolite accounts for about two thirds of the dose. In one study, rufinamide did not appear to significantly affect the plasma concentrations of other drugs used for the Lennox-Gastaut syndrome such as topiramate, lamotrigine, or valproic acid, but conflicting data suggest more robust interactions with other AEDs, including effects on rufinamide levels, especially in children.

STIRIPENTOL Stiripentol, though not a new molecule, was approved in Europe in 2007 for a very specific type of epilepsy. The drug is used with clobazam and valproate in the adjunctive therapy of refractory generalized tonic-clonic seizures in patients with severe myoclonic epilepsy of infancy (SMEI, Dravet’s syndrome) whose seizures are not adequately controlled with clobazam and valproate. The drug is legally imported into the USA on a compassionate use basis. The mechanism of action of stiripentol is not well understood but it has been shown to enhance GABAergic transmission in the brain, partly through a barbiturate-like effect, ie, prolonged opening of the Cl– channels in GABAA receptors. It also increases GABA levels in the brain. It can increase the effect of other AEDs by slowing their inactivation by cytochrome P450. Stiripentol is a potent inhibitor of CYP3A4, CYP1A2, and CYP2C19. Adverse effects of stiripentol itself are few, but the drug can dramatically increase the levels of valproate, clobazam, and the active metabolite of the latter, norclobazam. These drugs must be used cautiously together to avoid adverse effects. Dosing is complex, typically beginning with a reduction of the concomitant medication; stiripentol is then started at 10 mg/kg/d and is increased gradually to tolerability or to much higher doses. The kinetics of stiripentol are nonlinear.

TIAGABINE Tiagabine is a derivative of nipecotic acid and was “rationally designed” as an inhibitor of GABA uptake (as opposed to discovery through random screening).

Mechanism of Action Tiagabine is an inhibitor of GABA uptake in both neurons and glia. It preferentially inhibits the transporter isoform 1 (GAT-1) rather than GAT-2 or GAT-3 and increases extracellular GABA levels in the forebrain and hippocampus where GAT-1 is preferentially expressed. It prolongs the inhibitory action of synaptically released GABA, but its most significant effect may be potentiation of tonic inhibition. In rodents, it is potent against kindled seizures but weak against the maximal electroshock model, consistent with its predominant action in the forebrain and hippocampus.

Clinical Uses Tiagabine is indicated for the adjunctive treatment of partial seizures and is effective in doses ranging from 16 to 56 mg/d. Divided doses


as often as four times daily are sometimes required. Minor adverse events are dose related and include nervousness, dizziness, tremor, difficulty in concentrating, and depression. Excessive confusion, somnolence, or ataxia may require discontinuation. Psychosis occurs rarely. The drug can cause seizures in some patients, notably those taking the drug for other indications. Rash is an uncommon idiosyncratic adverse effect.

Pharmacokinetics Tiagabine is 90–100% bioavailable, has linear kinetics, and is highly protein bound. The half-life is 5–8 hours and decreases in the presence of enzyme-inducing drugs. Food decreases the peak plasma concentration but not the area under the concentration curve (see Chapter 3). Hepatic impairment causes a slight decrease in clearance and may necessitate a lower dose. The drug is oxidized in the liver by CYP3A. Elimination is primarily in the feces (60–65%) and urine (25%).

TOPIRAMATE Topiramate is a substituted monosaccharide that is structurally different from all other antiseizure drugs.

Mechanism of Action Topiramate blocks repetitive firing of cultured spinal cord neurons, as do phenytoin and carbamazepine. Its mechanism of action, therefore, is likely to involve blocking of voltage-gated Na+ channels. It also acts on high-voltage activated (L-type) Ca2+ channels. Topiramate potentiates the inhibitory effect of GABA, acting at a site different from the benzodiazepine or barbiturate sites. Topiramate also depresses the excitatory action of kainate on glutamate receptors. The multiple effects of topiramate may arise through a primary action on kinases altering the phosphorylation of voltage-gated and ligand-gated ion channels.

Clinical Uses Clinical trials of topiramate as monotherapy demonstrated efficacy against partial and generalized tonic-clonic seizures. The drug is also approved for the Lennox-Gastaut syndrome, and may be effective in infantile spasms and even absence seizures. Topiramate is also approved for the treatment of migraine headaches. The use of the drug in psychiatric disorders is controversial; convincing controlled data are lacking. Dosages typically range from 200 to 600 mg/d, with a few patients tolerating dosages higher than 1000 mg/d. Most clinicians begin at a low dose (50 mg/d) and increase slowly to prevent adverse effects. Several studies have used topiramate in monotherapy with encouraging results. Although no idiosyncratic reactions have been noted, dose-related adverse effects occur most frequently in the first 4 weeks and include somnolence, fatigue, dizziness, cognitive slowing, paresthesias, nervousness, and confusion. Acute myopia and glaucoma may require prompt drug withdrawal. Urolithiasis has also been reported. The drug is teratogenic in animal models, and hypospadias has been reported in male infants exposed in utero to topiramate; no causal relationship, however, could be established.

Pharmacokinetics Topiramate is rapidly absorbed (about 2 hours) and is 80% bioavailable. There is no food effect on absorption, minimal (15%) plasma protein binding, and only moderate (20–50%) metabolism; no active metabolites are formed. The drug is primarily excreted unchanged in the urine. The half-life is 20–30 hours. An extended-release formulation is available, which is promoted for once-daily administration. Although increased levels are seen with renal failure and hepatic impairment, there is no age or gender effect, no autoinduction, no inhibition of metabolism, and kinetics are linear. Drug interactions do occur and can be complex, but the major effect is on topiramate levels rather than on the levels of other antiseizure drugs. Birth control pills may be less effective in the presence of topiramate, and higher estrogen doses may be required.


VIGABATRIN Current investigations that seek drugs to enhance the effects of GABA include efforts to find GABA agonists and prodrugs, GABA transaminase inhibitors, and GABA uptake inhibitors. Vigabatrin is one such drug.

Mechanism of Action Vigabatrin is an irreversible inhibitor of GABA aminotransferase (GABA-T), the enzyme responsible for the degradation of GABA. It may also inhibit the vesicular GABA transporter. Vigabatrin produces a sustained increase in the extracellular concentration of GABA in the brain. This leads to some desensitization of synaptic GABAA receptors but prolonged activation of nonsynaptic GABAA receptors that provide tonic inhibition. A decrease in brain glutamine synthetase activity is probably secondary to the increased GABA concentrations. It is effective in a wide range of seizure models. Vigabatrin is marketed as a racemate; the S(+) enantiomer is active and the R(−) enantiomer appears to be inactive.

Clinical Uses Vigabatrin is useful in the treatment of partial seizures and infantile spasms. The half-life is approximately 6–8 hours, but considerable evidence suggests that the pharmacodynamic activity of the drug is more prolonged and not well correlated with the plasma half-life. In infants, the dosage is 50–150 mg/d. In adults, vigabatrin should be started at an oral dosage of 500 mg twice daily; a total of 2–3 g (rarely more) daily may be required for full effectiveness. Typical toxicities include drowsiness, dizziness, and weight gain. Less common but more troublesome adverse reactions are agitation, confusion, and psychosis; preexisting mental illness is a relative contraindication. The drug was delayed in its worldwide introduction by the appearance in rats and dogs of a reversible intramyelinic edema. This phenomenon has now been detected in infants taking the drug; the clinical significance is unknown. In addition, long-term therapy with vigabatrin has been associated with development of peripheral visual field defects in 30–50% of patients. The lesions are located in the retina, increase with drug exposure, and are usually not reversible. Newer techniques such as optical coherence tomography may better define the defect, which has proved difficult to quantify. Vigabatrin is usually reserved for use in patients with infantile spasms or with complex partial seizures refractory to other treatments.

ZONISAMIDE Zonisamide is a sulfonamide derivative. Its primary site of action appears to be the Na+ channel; it also acts on T-type voltage-gated Ca2+ channels. The drug is effective against partial and generalized tonic-clonic seizures and may also be useful against infantile spasms and certain myoclonias. It has good bioavailability, linear kinetics, low protein-binding, renal excretion, and a half-life of 1–3 days. Dosages range from 100 to 600 mg/d in adults and from 4 to 12 mg/d in children. Adverse effects include drowsiness, cognitive impairment, and potentially serious skin rashes. Zonisamide does not interact with other antiseizure drugs.

DRUGS USED IN GENERALIZED SEIZURES ETHOSUXIMIDE Ethosuximide was introduced in 1960 as the third of three marketed succinimides in the USA. Ethosuximide has very little activity against maximal electroshock but considerable efficacy against pentylenetetrazol seizures; it was introduced as a “pure petit mal” drug.

Chemistry Ethosuximide is the last antiseizure drug to be marketed whose origin is in the cyclic ureide structure. The three antiseizure succinimides marketed in the USA are ethosuximide, phensuximide, and methsuximide. Methsuximide and phensuximide have phenyl substituents, whereas ethosuximide is 2-ethyl-2-methylsuccinimide.


Mechanism of Action Ethosuximide has an important effect on Ca2+ currents, reducing the low-threshold (T-type) current. This effect is seen at therapeutically relevant concentrations in thalamic neurons. The T-type Ca 2+ currents are thought to provide a pacemaker current in thalamic neurons responsible for generating the rhythmic cortical discharge of an absence attack. Inhibition of this current could therefore account for the specific therapeutic action of ethosuximide. A recently described effect on inwardly rectifying K+ channels may also be significant.

Clinical Uses As predicted from its activity in laboratory models, ethosuximide is particularly effective against absence seizures, but has a very narrow spectrum of clinical activity. Documentation of its effectiveness in human absence seizures was achieved with long-term electroencephalographic recording techniques. Data continue to show that ethosuximide and valproate are the drugs of choice for absence seizures and are more effective than lamotrigine.

Pharmacokinetics Absorption is complete following administration of the oral dosage forms. Peak levels are observed 3–7 hours after oral administration of the capsules. Ethosuximide is not protein-bound. The drug is completely metabolized, principally by hydroxylation, to inactive metabolites. Ethosuximide has a very low total body clearance (0.25 L/kg/d). This corresponds to a half-life of approximately 40 hours, although values from 18 to 72 hours have been reported.

Therapeutic Levels & Dosage Therapeutic levels of 60–100 mcg/mL can be achieved in adults with dosages of 750–1500 mg/d, although lower or higher dosages and blood levels (up to 125 mcg/mL) may be necessary and tolerated in some patients. Ethosuximide has a linear relationship between dose and steady-state plasma levels. The drug might be administered as a single daily dose were it not for its adverse gastrointestinal effects; twice-a-day dosage is common.

Drug Interactions & Toxicity Administration of ethosuximide with valproic acid results in a decrease in ethosuximide clearance and higher steady-state concentrations owing to inhibition of metabolism. No other important drug interactions have been reported for the succinimides. The most common doserelated adverse effect of ethosuximide is gastric distress, including pain, nausea, and vomiting. When an adverse effect does occur, temporary dosage reductions may allow adaptation. Other dose-related adverse effects are transient lethargy or fatigue and, much less commonly, headache, dizziness, hiccup, and euphoria. Behavioral changes are usually in the direction of improvement. Non-dose-related or idiosyncratic adverse effects of ethosuximide are extremely uncommon.

PHENSUXIMIDE & METHSUXIMIDE Phensuximide (no longer readily available) and methsuximide are phenylsuccinimides that were developed and marketed before ethosuximide. They are used primarily as anti-absence drugs. Methsuximide is generally considered more toxic, and phensuximide less effective, than ethosuximide. Unlike ethosuximide, these two compounds have some activity against maximal electroshock seizures, and methsuximide has been used for partial seizures by some investigators.

VALPROIC ACID & SODIUM VALPROATE


Sodium valproate, also used as the free acid, valproic acid, was found to have antiseizure properties when used as a solvent in the search for other drugs effective against seizures. It was marketed in France in 1969 but was not licensed in the USA until 1978. Valproic acid is fully ionized at body pH, and for that reason the active form of the drug may be assumed to be the valproate ion regardless of whether valproic acid or a salt of the acid is administered.

Chemistry Valproic acid is one of a series of fatty carboxylic acids that have antiseizure activity; this activity appears to be greatest for carbon chain lengths of five to eight atoms. The amides and esters of valproic acid are also active antiseizure agents.

Mechanism of Action The time course of valproate’s anticonvulsant activity appears to be poorly correlated with blood or tissue levels of the parent drug, an observation giving rise to considerable speculation regarding both the active species and the mechanism of action of valproic acid. Valproate is active against both pentylenetetrazol and maximal electroshock seizures. Like phenytoin and carbamazepine, valproate blocks sustained high-frequency repetitive firing of neurons in culture at therapeutically relevant concentrations. Its action against partial seizures may be a consequence of this effect on Na+ currents. Blockade of NMDA receptor-mediated excitation may also be important. Much attention has been paid to the effects of valproate on GABA. Several studies have shown increased levels of GABA in the brain after administration of valproate, although the mechanism for this increase remains unclear. An effect of valproate to facilitate glutamic acid decarboxylase (GAD), the enzyme responsible for GABA synthesis, has been described. An inhibitory effect on the GABA transporter GAT-1 may contribute. At very high concentrations, valproate inhibits GABA transaminase in the brain, thus blocking degradation of GABA. However, at the relatively low doses of valproate needed to abolish pentylenetetrazol seizures, brain GABA levels may remain unchanged. Valproate produces a reduction in the aspartate content of rodent brain, but the relevance of this effect to its anticonvulsant action is not known. Valproic acid is a potent inhibitor of histone deacetylase and through this mechanism changes the transcription of many genes. A similar effect, but to a lesser degree, is shown by some other antiseizure drugs (topiramate, carbamazepine, and a metabolite of levetiracetam).

Clinical Uses Valproate is very effective against absence seizures and is often preferred to ethosuximide when the patient has concomitant generalized tonic-clonic attacks. Valproate is unique in its ability to control certain types of myoclonic seizures; in some cases the effect is very dramatic. The drug is effective in tonic-clonic seizures, especially those that are primarily generalized. A few patients with atonic attacks may also respond, and some evidence suggests that the drug is effective in partial seizures. Its use in epilepsy is at least as broad as that of any other drug. Intravenous formulations are occasionally used to treat status epilepticus. Other uses of valproate include management of bipolar disorder and migraine prophylaxis.

Pharmacokinetics Valproate is well absorbed after an oral dose, with bioavailability greater than 80%. Peak blood levels are observed within 2 hours. Food may delay absorption, and decreased toxicity may result if the drug is given after meals. Valproic acid is 90% bound to plasma proteins, although the fraction bound is somewhat reduced at blood levels greater than 150 mcg/mL. Since valproate is both highly ionized and highly protein-bound, its distribution is essentially confined to extracellular water, with a volume of distribution of approximately 0.15 L/kg. At higher doses, there is an increased free fraction of valproate, resulting in lower total drug levels than expected. It may be clinically useful, therefore, to measure both total and free drug levels. Clearance for valproate is low and dose dependent; its half-life varies from 9 to 18 hours. Approximately 20% of the drug is excreted as a direct conjugate of valproate. The sodium salt of valproate is marketed in Europe as a tablet and is quite hygroscopic. In Central and South America, the magnesium salt is available, which is considerably less hygroscopic. The free acid of valproate was first marketed in the USA in a capsule containing corn oil; the sodium salt is also available in syrup, primarily for pediatric use. An enteric-coated tablet of divalproex


sodium is also marketed in the USA. This improved product, a 1:1 coordination compound of valproic acid and sodium valproate, is as bioavailable as the capsule but is absorbed much more slowly and is preferred by many patients. Peak concentrations following administration of the enteric-coated tablets are seen in 3–4 hours. Various extended-release preparations are available; not all are bioequivalent and may require dosage adjustment.

Therapeutic Levels & Dosage Dosages of 25–30 mg/kg/d may be adequate in some patients, but others may require 60 mg/kg/d or even more. Therapeutic levels of valproate range from 50 to 100 mcg/mL.

Drug Interactions Valproate displaces phenytoin from plasma proteins. In addition to binding interactions, valproate inhibits the metabolism of several drugs, including phenobarbital, phenytoin, and carbamazepine, leading to higher steady-state concentrations of these agents. The inhibition of phenobarbital metabolism, for example, may cause levels of the barbiturate to rise steeply, causing stupor or coma. Valproate can dramatically decrease the clearance of lamotrigine.

Toxicity The most common dose-related adverse effects of valproate are nausea, vomiting, and other gastrointestinal complaints such as abdominal pain and heartburn. The drug should be started gradually to avoid these symptoms. Sedation is uncommon with valproate alone but may be striking when valproate is added to phenobarbital. A fine tremor is frequently seen at higher levels. Other reversible adverse effects, seen in a small number of patients, include weight gain, increased appetite, and hair loss. The idiosyncratic toxicity of valproate is largely limited to hepatotoxicity, but this may be severe; there seems little doubt that the hepatotoxicity of valproate has been responsible for more than 50 fatalities in the USA alone. The risk is greatest for patients under 2 years of age and for those taking multiple medications. Initial aspartate aminotransferase values may not be elevated in susceptible patients, although these levels do eventually become abnormal. Most fatalities have occurred within 4 months after initiation of therapy. Some clinicians recommend treatment with oral or intravenous L-carnitine as soon as severe hepatotoxicity is suspected. Careful monitoring of liver function is recommended when starting the drug; the hepatotoxicity is reversible in some cases if the drug is withdrawn. The other observed idiosyncratic response with valproate is thrombocytopenia, although documented cases of abnormal bleeding are lacking. It should be noted that valproate is an effective and popular antiseizure drug and that only a very small number of patients have had severe toxic effects from its use. Several epidemiologic studies of valproate have confirmed a substantial increase in the incidence of spina bifida in the offspring of women who took valproate during pregnancy. In addition, an increased incidence of cardiovascular, orofacial, and digital abnormalities has been reported. These observations must be strongly considered in the choice of drugs during pregnancy.

OXAZOLIDINEDIONES Trimethadione, the first oxazolidinedione (Figure 24–3), was introduced as an antiseizure drug in 1945 and remained the drug of choice for absence seizures until the introduction of succinimides in the 1950s. Use of the oxazolidinediones—trimethadione, paramethadione, and dimethadione—is now very limited; the latter two are not readily available. These compounds are active against pentylenetetrazol-induced seizures. Trimethadione raises the threshold for seizure discharges after repetitive thalamic stimulation. It—or, more notably, its active metabolite dimethadione—has the same effect on thalamic Ca 2+ currents as ethosuximide (reducing the T-type Ca 2+ current). Thus, suppression of absence seizures is likely to depend on inhibiting the pacemaker action of thalamic neurons. Trimethadione is rapidly absorbed, with peak levels reached within 1 hour after drug administration. It is not bound to plasma proteins. Trimethadione is completely metabolized in the liver by demethylation to dimethadione, which may exert the major antiseizure activity. Dimethadione has an extremely long half-life (240 hours). The therapeutic plasma level range for trimethadione has never been established, although trimethadione blood levels higher than 20 mcg/mL and dimethadione levels higher than 700 mcg/mL have been suggested. A dosage of 30 mg/kg/d of trimethadione is necessary to achieve these levels in adults. The most common and bothersome dose-related adverse effect of the oxazolidinediones is sedation. Trimethadione has been associated with many other toxic adverse effects, some of which are severe. These drugs should not be used during pregnancy.

OTHER DRUGS USED IN MANAGEMENT OF EPILEPSY Some drugs not classifiable by application to seizure type are discussed in this section.


BENZODIAZEPINES Six benzodiazepines play prominent roles in the therapy of epilepsy (see also Chapter 22). Although many benzodiazepines are similar chemically, subtle structural alterations result in differences in activity and pharmacokinetics. They have two mechanisms of antiseizure action, which are shown to different degrees by the six compounds. This is evident from the fact that diazepam is relatively more potent against electroshock and clonazepam against pentylenetetrazol (the latter effect correlating with an action at the GABA-benzodiazepine allosteric receptor sites). Possible mechanisms of action are discussed in Chapter 22. Diazepam given intravenously or rectally is highly effective for stopping continuous seizure activity, especially generalized tonicclonic status epilepticus (see below). The drug is occasionally given orally on a long-term basis, although it is not considered very effective in this application, probably because of the rapid development of tolerance. A rectal gel is available for refractory patients who need acute control of bouts of seizure activity. Lorazepam appears in some studies to be more effective and longer acting than diazepam in the treatment of status epilepticus and is preferred by some experts. Clonazepam is a long-acting drug with documented efficacy against absence seizures; on a milligram basis, it is one of the most potent antiseizure agents known. It is also effective in some cases of myoclonic seizures and has been tried in infantile spasms. Sedation is prominent, especially on initiation of therapy; starting doses should be small. Maximal tolerated doses are usually in the range of 0.1– 0.2 mg/kg, but many weeks of gradually increasing daily doses may be needed to achieve these dosages in some patients. Nitrazepam is not marketed in the USA but is used in many other countries, especially for infantile spasms and myoclonic seizures. It is less potent than clonazepam, and superiority to that drug has not been documented. Clorazepate dipotassium is approved in the USA as an adjunct to treatment of complex partial seizures in adults. Drowsiness and lethargy are common adverse effects, but as long as the drug is increased gradually, dosages as high as 45 mg/d can be given. Clobazam is widely used in a variety of seizure types. It is a 1,5-benzodiazepine (other marketed benzodiazepines are 1,4benzodiazepines) and reportedly has less sedative potential. Whether the drug has significant clinical advantages is not clear. It has a half-life of 18 hours and is effective at dosages of 0.5–1 mg/kg/d. It does interact with some other antiseizure drugs and causes adverse effects typical of the benzodiazepines; efficacy, in some patients, is limited by the development of tolerance. It has an active metabolite, norclobazam. The drug is approved in the USA for treatment of Lennox-Gastaut syndrome.

Pharmacokinetics See Chapter 22.

Limitations Two prominent aspects of benzodiazepines limit their usefulness. The first is their pronounced sedative effect, which is unfortunate both in the treatment of status epilepticus and in chronic therapy. Children may manifest a paradoxical hyperactivity, as with barbiturates. The second problem is tolerance, in which seizures may respond initially but recur within a few months. The remarkable antiseizure potency of these compounds often cannot be realized because of these limiting factors.

ACETAZOLAMIDE Acetazolamide is a diuretic whose main action is the inhibition of carbonic anhydrase (see Chapter 15). Mild acidosis in the brain may be the mechanism by which the drug exerts its antiseizure activity; alternatively, the depolarizing action of bicarbonate ions moving out of neurons via GABA receptor ion channels may be diminished by carbonic anhydrase inhibition. Acetazolamide has been used for all types of seizures but is severely limited by the rapid development of tolerance, with return of seizures usually within a few weeks. The drug may have a special role in epileptic women who experience seizure exacerbations at the time of menses; seizure control may be improved and tolerance may not develop because the drug is not administered continuously. The usual dosage is approximately 10 mg/kg/d to a maximum of 1000 mg/d. Another carbonic anhydrase inhibitor, sulthiame, was not found to be effective as an anticonvulsant in clinical trials in the USA. It is marketed in a number of other countries.

CLINICAL PHARMACOLOGY OF ANTISEIZURE DRUGS SEIZURE CLASSIFICATION In general, the type of medication used for epilepsy depends on the empiric nature of the seizure. For this reason, considerable effort has been expended to classify seizures so that clinicians will be able to make a “seizure diagnosis” and on that basis prescribe appropriate therapy. Errors in seizure diagnosis cause use of the wrong drugs, and an unpleasant cycle ensues in which poor seizure control is


followed by increasing drug doses and medication toxicity. As noted, seizures are divided into two groups: partial and generalized. Drugs used for partial seizures are more or less the same for all subtypes of partial seizures, but drugs used for generalized seizures are determined by the individual seizure subtype. A summary of the most widely used international classification of epileptic seizures is presented in Table 24–1.

Partial (Focal) Seizures Partial seizures are those in which a localized onset of the attack can be ascertained, either by clinical observation or by electroencephalographic recording; the attack begins in a specific locus in the brain. There are three types of partial seizures, determined to some extent by the degree of brain involvement by the abnormal discharge. The least complicated partial seizure is the simple partial seizure, characterized by minimal spread of the abnormal discharge such that normal consciousness and awareness are preserved. For example, the patient may have a sudden onset of clonic jerking of an extremity lasting 60–90 seconds; residual weakness may last for 15–30 minutes after the attack. The patient is completely aware of the attack and can describe it in detail. The electroencephalogram may show an abnormal discharge highly localized to the involved portion of the brain. The complex partial seizure also has a localized onset, but the discharge becomes more widespread (usually bilateral) and almost always involves the limbic system. Most complex partial seizures arise from one of the temporal lobes, possibly because of the susceptibility of this area of the brain to insults such as hypoxia or infection. Clinically, the patient may have a brief warning followed by an alteration of consciousness during which some patients stare and others stagger or even fall. Most, however, demonstrate fragments of integrated motor behavior called automatisms for which the patient has no memory. Typical automatisms are lip smacking, swallowing, fumbling, scratching, and even walking about. After 30–120 seconds, the patient makes a gradual recovery to normal consciousness but may feel tired or ill for several hours after the attack. The last type of partial seizure is the secondarily generalized attack, in which a partial seizure immediately precedes a generalized tonic-clonic (grand mal) seizure. This seizure type is described in the text that follows.

Generalized Seizures Generalized seizures are those in which there is no evidence of localized onset. The group is quite heterogeneous. Generalized tonic-clonic (grand mal) seizures are the most dramatic of all epileptic seizures and are characterized by tonic rigidity of all extremities, followed in 15–30 seconds by a tremor that is actually an interruption of the tonus by relaxation. As the relaxation phases become longer, the attack enters the clonic phase, with massive jerking of the body. The clonic jerking slows over 60–120 seconds, and the patient is usually left in a stuporous state. The tongue or cheek may be bitten, and urinary incontinence is common. Primary generalized tonic-clonic seizures begin without evidence of localized onset, whereas secondary generalized tonic-clonic seizures are preceded by another seizure type, usually a partial seizure. The medical treatment of both primary and secondary generalized tonicclonic seizures is the same and uses drugs appropriate for partial seizures. The absence (petit mal) seizure is characterized by both sudden onset and abrupt cessation. Its duration is usually less than 10 seconds and rarely more than 45 seconds. Consciousness is altered; the attack may also be associated with mild clonic jerking of the eyelids or extremities, with postural tone changes, autonomic phenomena, and automatisms. The occurrence of automatisms can complicate the clinical differentiation from complex partial seizures in some patients. Absence attacks begin in childhood or adolescence and may occur up to hundreds of times a day. The electroencephalogram during the seizure shows a highly characteristic 2.5–3.5 Hz spike-and-wave pattern. Atypical absence patients have seizures with postural changes that are more abrupt, and such patients are often mentally retarded; the electroencephalogram may show a slower spike-and-wave discharge, and the seizures may be more refractory to therapy. Myoclonic jerking is seen, to a greater or lesser extent, in a wide variety of seizures, including generalized tonic-clonic seizures, partial seizures, absence seizures, and infantile spasms. Treatment of seizures that include myoclonic jerking should be directed at the primary seizure type rather than at the myoclonus. Some patients, however, have myoclonic jerking as the major seizure type, and some have frequent myoclonic jerking and occasional generalized tonic-clonic seizures without overt signs of neurologic deficit. Many kinds of myoclonus exist, and much effort has gone into attempts to classify this entity. Atonic seizures are those in which the patient has sudden loss of postural tone. If standing, the patient falls suddenly to the floor and may be injured. If seated, the head and torso may suddenly drop forward. Although most often seen in children, this seizure type is not unusual in adults. Many patients with atonic seizures wear helmets to prevent head injury. Momentary increased tone may be observed in some patients, hence the use of the term “tonic-atonic seizure.” Infantile spasms are an epileptic syndrome and not a seizure type. The attacks, though sometimes fragmentary, are most often bilateral and are included for pragmatic purposes with the generalized seizures. These attacks are most often characterized clinically by brief, recurrent myoclonic jerks of the body with sudden flexion or extension of the body and limbs; the forms of infantile spasms are, however, quite heterogeneous. Ninety percent of affected patients have their first attack before the age of 1 year. Most patients are intellectually delayed, presumably from the same cause as the spasms. The cause is unknown in many patients, but such widely disparate


disorders as infection, kernicterus, tuberous sclerosis, and hypoglycemia have been implicated. In some cases, the electroencephalogram is characteristic. Drugs used to treat infantile spasms are effective only in some patients; there is little evidence that the cognitive retardation is alleviated by therapy, even when the attacks disappear.

THERAPEUTIC STRATEGY In designing a therapeutic strategy, the use of a single drug is preferred, especially in patients who are not severely affected and who can benefit from the advantage of fewer adverse effects using monotherapy. For patients with hard-to-control seizures, multiple drugs are usually utilized simultaneously. For most of the older antiseizure drugs, relationships between blood levels and therapeutic effects have been characterized to a high degree. The same is true for the pharmacokinetics of these drugs. These relationships provide significant advantages in the development of therapeutic strategies for the treatment of epilepsy. The therapeutic index for most antiseizure drugs is low, and toxicity is not uncommon. Thus, effective treatment of seizures often requires an awareness of the therapeutic levels and pharmacokinetic properties as well as the characteristic toxicities of each agent. Measurements of antiseizure drug plasma levels can be very useful when combined with clinical observations and pharmacokinetic data (Table 24–2). The relationship between seizure control and plasma drug levels is variable and often less clear for the drugs marketed since 1990. TABLE 24–2 Serum concentrations reference ranges for some antiseizure drugs.



MANAGEMENT OF EPILEPSY PARTIAL SEIZURES & GENERALIZED TONIC-CLONIC SEIZURES For many years, the choice of drugs for partial and tonic-clonic seizures was usually limited to phenytoin, carbamazepine, or barbiturates. There was a strong tendency to limit the use of sedative antiseizure drugs such as barbiturates and benzodiazepines to patients who could not tolerate other medications; this trend led, in the 1980s, to increased use of carbamazepine. Although carbamazepine and phenytoin remain widely used, most newer drugs (marketed after 1990) are effective against these same seizure types. With the older drugs, efficacy and long-term adverse effects are well established; this creates a confidence level in spite of questionable tolerability. Most newer drugs have a broader spectrum of activity, and many are well tolerated; therefore, the newer drugs are often preferred to the older ones. Although some data suggest that most of these newer drugs confer an increased risk of nontraumatic fractures, choosing a drug on this basis is not yet practical.

GENERALIZED SEIZURES The issues (described above) related to choosing between old and new drugs apply to the generalized group of seizures as well. The drugs used for generalized tonic-clonic seizures are the same as for partial seizures; in addition, valproate is clearly useful. At least three drugs are effective against absence seizures. Two are nonsedating and therefore preferred: ethosuximide and valproate. Clonazepam is also highly effective but has disadvantages of dose-related adverse effects and development of tolerance. Lamotrigine and topiramate may also be useful. Specific myoclonic syndromes are usually treated with valproate; an intravenous formulation can be used acutely if needed. It is nonsedating and can be dramatically effective. Other patients respond to clonazepam, nitrazepam, or other benzodiazepines, although high doses may be necessary, with accompanying drowsiness. Zonisamide and levetiracetam may be useful. Another specific myoclonic syndrome, juvenile myoclonic epilepsy, can be aggravated by phenytoin or carbamazepine; valproate is the drug of choice followed by lamotrigine and topiramate. Atonic seizures are often refractory to all available medications, although some reports suggest that valproate may be beneficial, as may lamotrigine. Benzodiazepines have been reported to improve seizure control in some of these patients but may worsen the attacks in others. Felbamate has been demonstrated to be effective in some patients, although the drug’s idiosyncratic toxicity limits its use. If the loss of tone appears to be part of another seizure type (eg, absence or complex partial seizures), every effort should be made to treat the other seizure type vigorously, hoping for simultaneous alleviation of the atonic component of the seizure. The ketogenic (high fat) diet may also be useful.

DRUGS USED IN INFANTILE SPASMS The treatment of infantile spasms is unfortunately limited to improvement of control of the seizures rather than other features of the disorder, such as retardation. Most patients receive a course of intramuscular corticotropin, although some clinicians note that prednisone may be equally effective and can be given orally. Clinical trials have been unable to settle the matter. In either case, therapy must often be discontinued because of adverse effects. If seizures recur, repeat courses of corticotropin or corticosteroids can be given, or other drugs may be tried. A repository corticotropin for injection is now approved in the USA for the treatment of infantile spasms, either of cryptogenic or symptomatic etiology. Other drugs widely used are the benzodiazepines such as clonazepam or nitrazepam; their efficacy in this heterogeneous syndrome may be nearly as good as that of corticosteroids. Vigabatrin is effective and is considered the drug of choice by many pediatric neurologists. The mechanism of action of corticosteroids or corticotropin in the treatment of infantile spasms is unknown but may involve reduction in inflammatory processes.

STATUS EPILEPTICUS There are many forms of status epilepticus. The most common, generalized tonic-clonic status epilepticus, is a life-threatening emergency, requiring immediate cardiovascular, respiratory, and metabolic management as well as pharmacologic therapy. The latter virtually always requires intravenous administration of antiseizure medications. Diazepam is the most effective drug in most patients for stopping the attacks and is given directly by intravenous push to a maximum total dose of 20–30 mg in adults. Intravenous diazepam may depress respiration (less frequently, cardiovascular function), and facilities for resuscitation must be immediately at hand during its administration. The effect of diazepam is not lasting, but the 30- to 40-minute seizure-free interval allows more definitive therapy to be initiated. Some physicians prefer lorazepam, which is equivalent to diazepam in effect and perhaps somewhat longer acting. For patients who are not actually in the throes of a seizure, diazepam therapy can be omitted and the patient treated at once with a long-acting drug such as phenytoin.


Until the introduction of fosphenytoin, the mainstay of continuing therapy for status epilepticus was intravenous phenytoin, which is effective and nonsedating. It can be given as a loading dose of 13–18 mg/kg in adults; the usual error is to give too little. Administration should be at a maximum rate of 50 mg/min. It is safest to give the drug directly by intravenous push, but it can also be diluted in saline; it precipitates rapidly in the presence of glucose. Careful monitoring of cardiac rhythm and blood pressure is necessary, especially in elderly people. At least part of the cardiotoxicity is from the propylene glycol in which the phenytoin is dissolved. Fosphenytoin, which is freely soluble in intravenous solutions without the need for propylene glycol or other solubilizing agents, is a safer parenteral agent. Because of its greater molecular weight, this prodrug is two thirds to three quarters as potent as phenytoin on a milligram basis. In previously treated epileptic patients, the administration of a large loading dose of phenytoin may cause some dose-related toxicity such as ataxia. This is usually a relatively minor problem during the acute status episode and is easily alleviated by later adjustment of plasma levels. For patients who do not respond to phenytoin, phenobarbital can be given in large doses: 100–200 mg intravenously to a total of 400– 800 mg. Respiratory depression is a common complication, especially if benzodiazepines have already been given, and there should be no hesitation in instituting intubation and ventilation. Although other drugs such as lidocaine have been recommended for the treatment of generalized tonic-clonic status epilepticus, general anesthesia is usually necessary in highly resistant cases. For patients in absence status, benzodiazepines are still drugs of first choice. Rarely, intravenous valproate may be required.

SPECIAL ASPECTS OF THE TOXICOLOGY OF ANTISEIZURE DRUGS TERATOGENICITY The potential teratogenicity of antiseizure drugs is controversial and important. It is important because teratogenicity resulting from longterm drug treatment of millions of people throughout the world may have a profound effect even if the effect occurs in only a small percentage of cases. It is controversial because both epilepsy and antiseizure drugs are heterogeneous, and few epileptic patients who are not receiving these drugs are available for study. Furthermore, patients with severe epilepsy, in whom genetic factors rather than drug factors may be of greater importance in the occurrence of fetal malformations, are often receiving multiple antiseizure drugs in high doses. In spite of these limitations, it appears—from whatever cause—that children born to mothers taking antiseizure drugs have an increased risk, perhaps twofold, of congenital malformations. Phenytoin has been implicated in a specific syndrome called fetal hydantoin syndrome, although not all investigators are convinced of its existence and a similar syndrome has been attributed both to phenobarbital and to carbamazepine. Valproate, as noted above, has also been implicated in a specific malformation, spina bifida. It is estimated that a pregnant woman taking valproic acid or sodium valproate has a 1–2% risk of having a child with spina bifida. Topiramate has shown some teratogenicity in animal testing and, as noted earlier, in the human male fetus. In dealing with the clinical problem of a pregnant woman with epilepsy, most epileptologists agree that although it is important to minimize exposure to antiseizure drugs, both in numbers and dosages, it is also important not to allow maternal seizures to go unchecked.

WITHDRAWAL Withdrawal of antiseizure drugs, whether by accident or by design, can cause increased seizure frequency and severity. The two factors to consider are the effects of the withdrawal itself and the need for continued drug suppression of seizures in the individual patient. In many patients, both factors must be considered. It is important to note, however, that the abrupt discontinuance of antiseizure drugs ordinarily does not cause seizures in nonepileptic patients, provided that the drug levels are not above the usual therapeutic range when the drug is stopped. Some drugs are more easily withdrawn than others. In general, withdrawal of anti-absence drugs is easier than withdrawal of drugs needed for partial or generalized tonic-clonic seizures. Barbiturates and benzodiazepines are the most difficult to discontinue; weeks or months may be required, with very gradual dosage decrements, to accomplish their complete outpatient removal. Because of the heterogeneity of epilepsy, complete discontinuance of antiseizure drug administration is an especially difficult problem. If a patient is seizure-free for 3 or 4 years, a trial of gradual discontinuance is often warranted.

OVERDOSE Antiseizure drugs are central nervous system depressants but are rarely lethal. Very high blood levels are usually necessary before overdoses can be considered life-threatening. The most dangerous effect of antiseizure drugs after large overdoses is respiratory depression, which may be potentiated by other agents, such as alcohol. Treatment of antiseizure drug overdose is supportive; stimulants should not be used. Efforts to hasten removal of antiseizure drugs, such as alkalinization of the urine (phenytoin is a weak acid), are usually ineffective.


SUICIDALITY An FDA analysis of suicidal behavior during clinical trials of antiseizure drugs was carried out in 2008. The presence of either suicidal behavior or suicidal ideation was 0.37% in patients taking active drugs and 0.24% in patients taking placebo. This, according to one analyst, represents an additional 2 of 1000 patients with such thoughts or behaviors. It is noteworthy that, although the entire class may receive some changes in labeling, the odds ratios for carbamazepine and for valproate were less than 1, and no data were available for phenytoin. Whether this effect is real or inextricably associated with this serious, debilitating disorder—with its inherently high rate of suicidality—is unclear.

ANTISEIZURE DRUGS IN DEVELOPMENT Three potential new antiseizure drugs are in phase 2 or phase 3 development; these are brivaracitam, YKP3089, and ganaxolone. Other drugs are less advanced but can be found on the epilepsy website at http://www.epilepsy.com/etp/pipeline_new_therapies.

SUMMARY Antiseizure Drugs




PREPARATIONS AVAILABLE

REFERENCES Avorn J: Drug warnings that can cause fits—Communicating risks in a data-poor environment. N Engl J Med 2008;359:991. Bialer M: Progress report on new antiepileptic drugs: A summary of the tenth EILAT conference (EILAT X). Epilepsy Res 2010;92:89. Cross SA, Curran MP: Lacosamide in partial onset seizures. Drugs 2009;69:449. Edwards HB et al: Minimizing pharmacodynamic interactions of high doses of lacosamide. Acta Neurol Scand 2012;125:228. Ettinger AB, Argoff CE: Use of antiepileptic drugs for non-epileptic conditions: Psychiatric disorders and chronic pain. Neurotherapeutics 2007;4:75. Faught, E: Ezogabine: A new angle on potassium gates. Epilepsy Currents, 2011;11:75.


French JA et al: Historical control monotherapy design in the treatment of epilepsy. Epilepsia 2010:51:1936. French JA et al: Development of new treatment approaches for epilepsy: Unmet needs and opportunities. Epilepsia 2013;54 (Suppl 4):3. Glauser T A et al: Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy. N Engl J Med 2010;362:790. Kaminski RM et al: SV2A is a broad-spectrum anticonvulsant target: Functional correlation between protein binding and seizure protection in models of both partial and generalized epilepsy. Neuropharmacol 2008;54:715. Kobayashi T et al: Inhibitory effects of the antiepileptic drug ethosuximide on G-protein-activated inwardly rectifying K+ channels. Neuropharmacol 2009;56:499. Meldrum BS, Rogawski MA: Molecular targets for antiepileptic drug development. Neurotherapeutics 2007;4:18. Molgaard-Nielsen D, Hviid A: Newer-generation antiepileptic drugs and the risk of major birth defects. JAMA 2011;305:1996. Porter RJ et al: Clinical development of drugs for epilepsy: A review of approaches in the United States and Europe. Epilepsy Research 2010;89:163. Porter RJ et al: AED mechanisms and principles of drug treatment. In: Stefan H, T heodore W (editors): Handbook of Clinical Neurology, 3rd series, Epilepsies Part 2: Treatment. Elsevier, 2012. Rogawski MA, Hanada T : Preclinical pharmacology of perampanel, a selective non-competitive AMPA receptor antagonist. Acta Neurol Scand 2013;127 (Suppl 197):19. Steinhof BJ et al: Efficacy and safety of adjunctive perampanel for the treatment of refractory partial seizures: A pooled analysis of three phase III studies. Epilepsia 2013;54:1481. Wilcox KS et al: Issues related to development of new anti-seizure treatments. Epilepsia 2013;54 (Suppl 4):24. Wolff C et al: Drug binding assays do not reveal specific binding of lacosamide to collapsin response mediator protein 2 (CRMP-2). CNS Neurosci T her 2012;18:493.

CASE STUDY ANSWER Lamotrigine was gradually added to the regimen to a dosage of 200 mg bid. Since then, the patient has been seizure-free for almost 2 years but now comes to the office for a medication review. Gradual discontinuation of levetiracetam is planned if the patient continues to do well for another year, although risk of recurrent seizures is always present when medications are withdrawn.


CHAPTER

25 General Anesthetics Helge Eilers, MD, & Spencer Yost, MD

CASE STUDY An elderly man with type 2 diabetes mellitus and ischemic pain in the lower extremity is scheduled for femoral-to-popliteal bypass surgery. He has a history of hypertension and coronary artery disease with symptoms of stable angina and can walk only half a block before pain in his legs forces him to stop. He has a 50-pack-a-year smoking history but stopped 2 years ago. His medications include atenolol, atorvastatin, and hydrochlorothiazide. The nurse in the preoperative holding area obtains the following vital signs: temperature 36.8°C (98.2°F), blood pressure 168/100 mm Hg, heart rate 78 bpm, oxygen saturation by pulse oximeter 96% while breathing room air, pain 5/10 in the right lower leg. What anesthetic agents will you choose and why? Does the choice of anesthetic make a difference?

For centuries, humankind has relied on natural medicines and physical methods to control surgical pain. Historical texts describe the sedative effects of cannabis, henbane, mandrake, and opium poppy. Physical methods such as cold, nerve compression, carotid artery occlusion, and cerebral concussion were also employed, with variable effect. Although surgery was performed under ether anesthesia as early as 1842, the first public demonstration of surgical general anesthesia in 1846 is generally accepted as the start of the modern era of anesthesia. For the first time physicians had a reliable means to keep their patients from experiencing pain during surgical procedures. The neurophysiologic state produced by general anesthetics is characterized by five primary effects: unconsciousness, amnesia, analgesia, inhibition of autonomic reflexes, and skeletal muscle relaxation. None of the currently available anesthetic agents when used alone can achieve all five of these desired effects well. In addition, an ideal anesthetic drug should induce rapid, smooth loss of consciousness, be rapidly reversible upon discontinuation, and possess a wide margin of safety. The modern practice of anesthesiology relies on the use of combinations of intravenous and inhaled drugs (balanced anesthesia techniques) to take advantage of the favorable properties of each agent while minimizing their adverse effects. The choice of anesthetic technique is determined by the type of diagnostic, therapeutic, or surgical intervention to be performed. For minor superficial surgery or for invasive diagnostic procedures, oral or parenteral sedatives can be used in combination with local anesthetics, so-called monitored anesthesia care techniques (see Box: Sedation & Monitored Anesthesia Care, and Chapter 26). These techniques provide profound analgesia, with retention of the patient’s ability to maintain a patent airway and to respond to verbal commands. For more extensive surgical procedures, anesthesia may begin with preoperative benzodiazepines, be induced with an intravenous agent (eg, thiopental or propofol), and be maintained with a combination of inhaled (eg, volatile agents, nitrous oxide) or intravenous drugs (eg, propofol, opioid analgesics), or both.

MECHANISM OF GENERAL ANESTHETIC ACTION General anesthetics have been in clinical use for more than 160 years but their mechanism of action remains unknown. Initial research focused on identifying a single biologic site of action for these drugs. In recent years this “unitary theory” of anesthetic action has been supplanted by a more complex picture of molecular targets located at multiple levels of the central nervous system (CNS). Anesthetics affect neurons at various cellular locations, but the primary focus has been on the synapse. A presynaptic action may alter the release of neurotransmitters, whereas a postsynaptic effect may change the frequency or amplitude of impulses exiting the synapse. At the organ level, the effect of anesthetics may result from strengthening inhibition or from diminishing excitation within the CNS. Studies on isolated spinal cord tissue have demonstrated that excitatory transmission is impaired more strongly by anesthetics than inhibitory effects are potentiated.


Sedation & Monitored Anesthesia Care Many diagnostic and minor therapeutic surgical procedures can be performed without general anesthesia using sedation-based anesthetic techniques. In this setting, regional or local anesthesia supplemented with midazolam or propofol and opioid analgesics (or ketamine) may be a more appropriate and safer approach than general anesthesia for superficial surgical procedures. This anesthetic technique is known as monitored anesthesia care, often abbreviated as MAC, not to be confused with the minimal alveolar concentration for the comparison of potencies of inhaled anesthetics (see text and Box: What Does Anesthesia Represent & Where Does It Work?). The technique typically involves the use of intravenous midazolam for premedication (to provide anxiolysis, amnesia, and mild sedation) followed by a titrated, variable-rate propofol infusion (to provide moderate to deep levels of sedation). A potent opioid analgesic or ketamine may be added to minimize the discomfort associated with the injection of local anesthesia and the surgical manipulations. Another approach, used primarily by nonanesthesiologists, is called conscious sedation. This technique refers to drug-induced alleviation of anxiety and pain in combination with an altered level of consciousness associated with the use of smaller doses of sedative medications. In this state the patient retains the ability to maintain a patent airway and is responsive to verbal commands. A wide variety of intravenous anesthetic drugs have proved to be useful drugs in conscious sedation techniques (eg, diazepam, midazolam, propofol). Use of benzodiazepines and opioid analgesics (eg, fentanyl) in conscious sedation protocols has the advantage of being reversible by the specific receptor antagonist drugs (flumazenil and naloxone, respectively). A specialized form of sedation is occasionally required in the intensive care unit (ICU), when patients are under severe stress and require mechanical ventilation for prolonged periods. In this situation, sedative-hypnotic drugs and low doses of intravenous anesthetics may be combined. Recently, dexmedetomidine has become a popular choice for this indication. Deep sedation is similar to a light state of general anesthesia characterized by decreased consciousness from which the patient is not easily aroused. The transition from deep sedation to general anesthesia is fluid and can be difficult to define. Because deep sedation is often accompanied by a loss of protective reflexes, an inability to maintain a patent airway and lack of verbal responsiveness to surgical stimuli, this state may be indistinguishable from general anesthesia. A practitioner with expertise in airway management, such as an anesthesiologist or nurse anesthetist, must be present. Intravenous agents used in deep sedation protocols mainly include the sedative-hypnotics propofol and midazolam, sometimes in combination with potent opioid analgesics or ketamine, depending on the level of pain associated with the surgery or procedure. Chloride channels (γ-aminobutyric acid-A [GABA A] and glycine receptors) and potassium channels (K2P , possibly KV, and KAT P channels) remain the primary inhibitory ion channels considered legitimate candidates of anesthetic action. Excitatory ion channel targets include those activated by acetylcholine (nicotinic and muscarinic receptors), by glutamate (amino-3-hydroxy-5-methyl-4-isoxazolpropionic acid [AMPA], kainate, and N-methyl-D-aspartate [NMDA] receptors), or by serotonin (5-HT2 and 5-HT3 receptors). Figure 25–1 depicts the relation of these inhibitory and excitatory targets of anesthetics within the context of the nerve terminal.



FIGURE 25–1 Putative targets of anesthetic action. Anesthetic drugs may (A) enhance inhibitory synaptic activity or (B) diminish excitatory activity. ACh, acetylcholine; GABAA, γ-aminobutyric acid-A.

INHALED ANESTHETICS A clear distinction should be made between volatile and gaseous anesthetics, both of which are administered by inhalation. Volatile anesthetics (halothane, enflurane, isoflurane, desflurane, sevoflurane) have low vapor pressures and thus high boiling points so that they are liquids at room temperature (20°C) and sea-level ambient pressure, whereas gaseous anesthetics (nitrous oxide, xenon) have high vapor pressures and low boiling points such that they are in gas form at room temperature. The special characteristics of volatile anesthetics make it necessary that they be administered using vaporizers. Figure 25–2 shows the chemical structures of important, clinically used, inhaled anesthetics.


FIGURE 25–2 Chemical structures of inhaled anesthetics.

PHARMACOKINETICS Inhaled anesthetics, volatile as well as gaseous, are taken up through gas exchange in the alveoli of the lung. Uptake from the alveoli into the blood and distribution and partitioning into the effect compartments are important determinants of the kinetics of these agents. As previously mentioned, an ideal anesthetic should have a rapid onset (induction), and its effect should be rapidly terminated. To achieve this, the effect site concentration in the CNS (brain and spinal cord) will need to change rapidly. Several factors determine how quickly the CNS concentration changes.


Uptake & Distribution A. Factors Controlling Uptake 1. Inspired concentration and ventilation—The driving force for uptake of an inhaled anesthetic into the body is the alveolar concentration. Two parameters that can be controlled by the anesthesiologist determine how quickly the alveolar concentration changes: (1) inspired concentration or partial pressure , and (2) alveolar ventilation. The partial pressure of an inhaled anesthetic in the inspired gas mixture directly affects the maximum partial pressure that can be achieved in the alveoli as well as the rate of increase of the partial pressure in the alveoli. Increases in the inspired partial pressure increase the gradient between inspired and alveolar partial pressure to accelerate induction. The increase of partial pressure in the alveoli is usually expressed as a ratio of alveolar concentration (FA) over inspired concentration (FI); the faster FA/FI approaches 1 (representing inspired-to-alveolar equilibrium), the faster anesthesia will occur during an inhaled induction. The primary parameter other than inspired concentration that directly controls the rate by which FA/FI approaches 1 is alveolar ventilation. An increase in ventilation will increase the rate of rise. The magnitude of the effect varies according to the blood:gas partition coefficient. An increase in pulmonary ventilation is accompanied by only a slight increase in arterial tension of an anesthetic with low blood solubility, but can significantly increase tension of agents with moderate to high blood solubility (Figure 25–3). For example, a fourfold increase in the ventilation rate almost doubles the FA/FI ratio for halothane during the first 10 minutes of administration but increases the FA/FI ratio for nitrous oxide by only 15%. Thus, hyperventilation increases the speed of induction of anesthesia with inhaled anesthetics that would normally have a slow onset. Depression of respiration by opioid analgesics slows the onset of anesthesia of inhaled anesthetics unless ventilation is manually or mechanically assisted.

FIGURE 25–3 Effect of ventilation on FA/FI and induction of anesthesia. Increased ventilation (8 L/min versus 2 L/min) accelerates the rate of rise toward equilibration of both halothane and nitrous oxide but results in a larger percentage increase for halothane in the first few minutes of induction. 2. Solubility—As described above, the rate of rise of FA/FI is an important determinant of the speed of induction, but is opposed by the uptake of anesthetic into the blood. Uptake is determined by pharmacokinetic characteristics of each anesthetic agent as well as patient factors. One of the most important factors influencing the transfer of an anesthetic from the lungs to the arterial blood is its solubility characteristics (Table 25–1). The blood:gas partition coefficient is a useful index of solubility and defines the relative affinity of an


anesthetic for the blood compared with that of inspired gas. The partition coefficients for desflurane and nitrous oxide, which are relatively insoluble in blood, are extremely low. When an anesthetic with low blood solubility diffuses from the lung into the arterial blood, relatively few molecules are required to raise its partial pressure; therefore, the arterial tension rises rapidly (Figure 25–4, top; nitrous oxide, desflurane, sevoflurane). Conversely, for anesthetics with moderate to high solubility (Figure 25–4, bottom; halothane, isoflurane), more molecules dissolve in the blood before partial pressure changes significantly, and arterial tension of the gas increases less rapidly. A blood:gas partition coefficient of 0.47 for nitrous oxide means that at equilibrium, the concentration in blood is less than half the concentration in the alveolar space (gas). A larger blood:gas partition coefficient produces a greater uptake of anesthetic and therefore increases the time required for FA/FI to approach equilibrium (Figure 25–4). TABLE 25–1 Pharmacologic properties of inhaled anesthetics.


FIGURE 25–4 The alveolar anesthetic concentration (FA) approaches the inspired anesthetic concentration (FI) fastest for the least soluble agents. 3. Cardiac output—Changes in pulmonary blood flow have obvious effects on the uptake of anesthetic gases from the alveolar space. An increase in pulmonary blood flow (ie, increased cardiac output) will increase the uptake of anesthetic, thereby decreasing the rate by which FA/FI rises, which will decrease the rate of induction of anesthesia. To better understand this mechanism, one should think about the effect of cardiac output in combination with the tissue distribution and uptake of anesthetic into other tissue compartments. An increase in cardiac output and pulmonary blood flow will increase uptake of anesthetic into the blood, but the anesthetic taken up will be distributed in all tissues, not just the CNS. Cerebral blood flow is well regulated and the increased cardiac output will therefore increase delivery of anesthetic to other tissues and not the brain. 4. Alveolar-venous partial pressure difference—The anesthetic partial pressure difference between alveolar and mixed venous blood is dependent mainly on uptake of the anesthetic by the tissues, including nonneural tissues. Depending on the rate and extent of tissue uptake, venous blood returning to the lungs may contain significantly less anesthetic than arterial blood. The greater this difference in anesthetic gas tensions, the more time it will take to achieve equilibrium with brain tissue. Anesthetic uptake into tissues is influenced by factors similar to those that determine transfer of the anesthetic from the lung to the intravascular space, including tissue:blood partition coefficients, rates of blood flow to the tissues, and concentration gradients. During the induction phase of anesthesia (and the initial phase of the maintenance period), the tissues that exert greatest influence on the arteriovenous anesthetic concentration gradient are those that are highly perfused (eg, brain, heart, liver, kidneys, and splanchnic bed). Combined, these tissues receive over 75% of the resting cardiac output. In the case of volatile anesthetics with relatively high solubility in highly perfused tissues, venous blood concentration initially is very low, and equilibrium with the alveolar space is achieved slowly. During maintenance of anesthesia with inhaled anesthetics, the drug continues to be transferred between various tissues at rates dependent on the solubility of the agent, the concentration gradient between the blood and the tissue, and the tissue blood flow. Although muscle and skin constitute 50% of the total body mass, anesthetics accumulate more slowly in these tissues than in highly perfused


tissues (eg, brain) because they receive only one fifth of the resting cardiac output. Although most anesthetic agents are highly soluble in adipose (fatty) tissues, the relatively low blood perfusion to these tissues delays accumulation, and equilibrium is unlikely to occur with most anesthetics during a typical 1- to 3-hour operation. The combined effect of ventilation, solubility in the different tissues, cardiac output, and blood flow distribution determines the rate of rise of FA/FI characteristic of each drug. Figure 25–5 schematically compares how uptake and distribution proceeds with two widely different agents. The anesthetic state is achieved when the partial pressure of the anesthetic in the brain reaches a threshold concentration determined by its potency (MAC; see Table 25–1 and Box: What Does Anesthesia Represent & Where Does It Work?). For an insoluble agent like desflurane the alveolar partial pressure can quickly equilibrate through the blood and brain compartments to reach anesthetizing concentrations. However, for an agent like halothane, its greater solubility in blood and other tissue compartments (higher partition coefficients) produce a steeper decline in the concentration gradient from lung to brain, causing a delayed onset of anesthesia. Therefore administering a larger concentration of halothane and increasing alveolar ventilation are the two strategies that can be used by anesthesiologists to speed the rate of induction with halothane.

FIGURE 25–5 Why induction of anesthesia is slower with more soluble anesthetic gases. In this schematic diagram, solubility in blood is represented by the relative size of the blood compartment (the more soluble, the larger the compartment). Relative partial pressures of the agents in the compartments are indicated by the degree of filling of each compartment. For a given concentration or partial pressure of the two anesthetic gases in the inspired air, it will take much longer for the blood partial pressure of the more soluble gas (halothane) to rise to the same partial pressure as in the alveoli. Since the concentration of the anesthetic agent in the brain can rise no faster than the concentration in the blood, the onset of anesthesia will be slower with halothane than with nitrous oxide. B. Elimination Recovery from inhalation anesthesia follows some of the same principles in reverse that are important during induction. The time to recovery from inhalation anesthesia depends on the rate of elimination of the anesthetic from the brain. One of the most important factors governing rate of recovery is the blood:gas partition coefficient of the anesthetic agent. Other factors controlling rate of recovery include pulmonary blood flow, magnitude of ventilation, and tissue solubility of the anesthetic. Two features differentiate the recovery phase from the induction phase. First, transfer of an anesthetic from the lungs to blood can be enhanced by increasing its concentration in inspired air, but the reverse transfer process cannot be enhanced because the concentration in the lungs cannot be reduced below zero. Second, at the beginning of the recovery phase, the anesthetic gas tension in different tissues may be quite variable, depending on the specific agent and the duration of anesthesia. In contrast, at the start of induction of anesthesia the initial anesthetic tension is zero in all tissues. Inhaled anesthetics that are relatively insoluble in blood (ie, possess low blood:gas partition coefficients) and brain are eliminated faster than the more soluble anesthetics. The washout of nitrous oxide, desflurane, and sevoflurane occurs at a rapid rate, leading to a more rapid recovery from their anesthetic effects compared with halothane and isoflurane. Halothane is approximately twice as soluble in brain tissue and five times more soluble in blood than nitrous oxide and desflurane; its elimination therefore takes place more slowly, and recovery from halothane- and isoflurane-based anesthesia is predictably less rapid.


The duration of exposure to the anesthetic can also have a significant effect on the recovery time, especially in the case of the more soluble anesthetics (eg, halothane and isoflurane). Accumulation of anesthetics in muscle, skin, and fat increases with prolonged exposure (especially in obese patients), and blood tension may decline slowly during recovery as the anesthetic is slowly eliminated from these tissues. Although recovery may be rapid even with the more soluble agents following a short period of exposure, recovery is slow after prolonged administration of halothane or isoflurane. 1. Ventilation—Two parameters that can be manipulated by the anesthesiologist are useful in controlling the speed of induction of and recovery from inhaled anesthesia: (1) concentration of anesthetic in the inspired gas and (2) alveolar ventilation. Because the concentration of anesthetic in the inspired gas cannot be reduced below zero, hyperventilation is the only way to speed recovery. 2. Metabolism—Modern inhaled anesthetics are eliminated mainly by ventilation and are only metabolized to a very small extent; thus, metabolism of these drugs does not play a significant role in the termination of their effect. However, metabolism may have important implications for their toxicity (see Toxicity of Anesthetic Agents). Hepatic metabolism may also contribute to the elimination of and recovery from some older volatile anesthetics. For example, halothane is eliminated more rapidly during recovery than enflurane, which would not be predicted from their respective tissue solubility. This increased elimination occurs because over 40% of inspired halothane is metabolized during an average anesthetic procedure, whereas less than 10% of enflurane is metabolized over the same period. In terms of the extent of hepatic metabolism, the rank order for the inhaled anesthetics is halothane > enflurane > sevoflurane > isoflurane > desflurane > nitrous oxide (Table 25–1). Nitrous oxide is not metabolized by human tissues. However, bacteria in the gastrointestinal tract may be able to break down the nitrous oxide molecule.

PHARMACODYNAMICS Organ System Effects of Inhaled Anesthetics A. Cerebral Effects Anesthetic potency is currently described by the minimal alveolar concentration (MAC) required to prevent a response to a surgical incision (see Box: What Does Anesthesia Represent & Where Does It Work?). Inhaled anesthetics (and intravenous anesthetics, discussed later) decrease the metabolic activity of the brain. Decreased cerebral metabolic rate (CMR) generally reduces blood flow within the brain. However, volatile anesthetics also cause cerebral vasodilation, which can increase cerebral blood flow. The net effect on cerebral blood flow (increase, decrease, or no change) depends on the concentration of anesthetic delivered. At 0.5 MAC, the reduction in CMR is greater than the vasodilation caused by the anesthetic, so cerebral blood flow is decreased. Conversely, at 1.5 MAC, vasodilation by the anesthetic is greater than the reduction in CMR, so cerebral blood flow is increased. In between, at 1.0 MAC, the effects are balanced and cerebral blood flow is unchanged. An increase in cerebral blood flow is clinically undesirable in patients who have increased intracranial pressure because of brain tumor, intracranial hemorrhage, or head injury. Therefore, administration of high concentrations of volatile anesthetics is undesirable in patients with increased intracranial pressure. Hyperventilation can be used to attenuate this response; decreasing the PaCO2 (the partial pressure of carbon dioxide in arterial blood) through hyperventilation causes cerebral vasoconstriction. If the patient is hyperventilated before the volatile agent is started, the increase in intracranial pressure can be minimized. Nitrous oxide can increase cerebral blood flow and cause increased intracranial pressure. This effect is most likely caused by activation of the sympathetic nervous system (as described below). Therefore, nitrous oxide may be combined with other agents (intravenous anesthetics) or techniques (hyperventilation) that reduce cerebral blood flow in patients with increased intracranial pressure.

What Does Anesthesia Represent & Where Does It Work? Anesthetic action has three principal components: immobility, amnesia, and unconsciousness. Immobility Immobility is the easiest anesthetic end point to measure. Edmond Eger and colleagues introduced the concept of minimal alveolar concentration (MAC) to quantify the potency of an inhalational anesthetic. They defined 1.0 MAC as the partial pressure of an inhalational anesthetic in the alveoli of the lungs at which 50% of a population of nonrelaxed patients remained immobile at the time of a skin incision. Anesthetic immobility is mediated primarily by neural inhibition within the spinal cord but may also include inhibited nociceptive transmission to the brain. Amnesia The ablation of memory arises from several locations in the CNS, including the hippocampus, amygdala, prefrontal cortex, and regions of the sensory and motor cortices. Memory researchers differentiate two types of memory: (1) explicit memory, ie, specific


awareness or consciousness under anesthesia; and (2) implicit memory, the unconscious acquisition of information under adequate levels of anesthesia. Their studies have found that formation of both types of memory is reliably prevented at low MAC values (0.2–0.4 MAC). Prevention of explicit memory (awareness) has spurred the development of monitors such as the bispectral index, electroencephalogram (EEG), and entropy monitor of auditory evoked potentials to help recognize inadequate planes of anesthesia. Consciousness The ability of anesthetic drugs to abolish consciousness requires action at anatomic locations responsible for the formation of human consciousness. Leading neuroscientists studying consciousness identify three regions in the brain involved in generating personal awareness: the cerebral cortex, the thalamus, and the reticular activating system. These regions seem to interact as a cortical system via identified pathways, producing a state in which humans are awake, aware, and perceiving. Our current state of understanding supports the following framework: sensory stimuli conducted through the reticular formation of the brainstem into supratentorial signaling loops, connecting the thalamus with various regions of the cortex, are the foundation of consciousness. These neural pathways involved in the development of consciousness are disrupted by anesthetics. Potent inhaled anesthetics produce a basic pattern of change to brain electrical activity as recorded by standard electroencephalography. Isoflurane, desflurane, sevoflurane, halothane, and enflurane produce initial activation of the EEG at low doses and then slowing of electrical activity up to doses of 1.0–1.5 MAC. At higher concentrations, EEG suppression increases to the point of electrical silence with isoflurane at 2.0–2.5 MAC. Isolated epileptic-like patterns may also be seen between 1.0 and 2.0 MAC, especially with sevoflurane and enflurane, but frank clinical seizure activity has been observed only with enflurane. Nitrous oxide used alone causes fast electrical oscillations emanating from the frontal cortex at doses associated with analgesia and depressed consciousness. Traditionally, anesthetic effects on the brain produce four stages or levels of increasing depth of CNS depression ( Guedel’s signs , derived from observations of the effects of inhaled diethyl ether): Stage I—analgesia: The patient initially experiences analgesia without amnesia. Later in stage I, both analgesia and amnesia are produced. Stage II—excitement: During this stage, the patient appears delirious, may vocalize but is completely amnesic. Respiration is rapid, and heart rate and blood pressure increase. Duration and severity of this light stage of anesthesia is shortened by rapidly increasing the concentration of the agent. Stage III—surgical anesthesia: This stage begins with slowing of respiration and heart rate and extends to complete cessation of spontaneous respiration (apnea). Four planes of stage III are described based on changes in ocular movements, eye reflexes, and pupil size, indicating increasing depth of anesthesia. Stage IV—medullary depression: This deep stage of anesthesia represents severe depression of the CNS, including the vasomotor center in the medulla and respiratory center in the brainstem. Without circulatory and respiratory support, death would rapidly ensue. B. Cardiovascular Effects Halothane, enflurane, isoflurane, desflurane, and sevoflurane all depress normal cardiac contractility (halothane and enflurane more so than isoflurane, desflurane, and sevoflurane). As a result, all volatile agents tend to decrease mean arterial pressure in direct proportion to their alveolar concentration. With halothane and enflurane, the reduced arterial pressure is caused primarily by myocardial depression (reduced cardiac output) and there is little change in systemic vascular resistance. In contrast, isoflurane, desflurane, and sevoflurane produce greater vasodilation with minimal effect on cardiac output. These differences may have important implications for patients with heart failure. Because isoflurane, desflurane, and sevoflurane better preserve cardiac output as well as reduce preload (ventricular filling) and afterload (systemic vascular resistance), these agents may be better choices for patients with impaired myocardial function. Nitrous oxide also depresses myocardial function in a concentration-dependent manner. This depression may be significantly offset by a concomitant activation of the sympathetic nervous system resulting in preservation of cardiac output. Therefore, administration of nitrous oxide in combination with the more potent volatile anesthetics can minimize circulatory depressant effects by both anestheticsparing and sympathetic-activating actions. Because all inhaled anesthetics produce a dose-dependent decrease in arterial blood pressure, activation of autonomic nervous system reflexes may trigger increased heart rate. However, halothane, enflurane, and sevoflurane have little effect on heart rate, probably because they attenuate baroreceptor input into the autonomic nervous system. Desflurane and isoflurane significantly increase heart rate because they cause less depression of the baroreceptor reflex. In addition, desflurane can trigger transient sympathetic activation—with elevated catecholamine levels—to cause marked increases in heart rate and blood pressure during administration of high desflurane concentrations or when desflurane concentrations are changed rapidly. Inhaled anesthetics tend to reduce myocardial oxygen consumption, which reflects depression of normal cardiac contractility and decreased arterial blood pressure. In addition, inhaled anesthetics produce coronary vasodilation. The net effect of decreased oxygen demand and increased coronary flow (oxygen supply) is improved myocardial oxygenation. However, other factors such as surgical stimulation, intravascular volume status, blood oxygen levels, and withdrawal of perioperative β blockers, may tilt the oxygen supplydemand balance toward myocardial ischemia. Halothane and, to a lesser extent, other volatile anesthetics sensitize the myocardium to epinephrine and circulating catecholamines. Ventricular arrhythmias may occur when patients under anesthesia with halothane are given sympathomimetic drugs or have high circulating levels of endogenous catecholamines (eg, anxious patients, administration of epinephrine-containing local anesthetics, inadequate intraoperative anesthesia or analgesia, patients with pheochromocytomas). This effect is less marked for isoflurane,


sevoflurane, and desflurane. C. Respiratory Effects All volatile anesthetics possess varying degrees of bronchodilating properties, an effect of value in patients with active wheezing and in status asthmaticus. However, airway irritation, which may provoke coughing or breath-holding, is induced by the pungency of some volatile anesthetics. The pungency of isoflurane and desflurane makes these agents less suitable for induction of anesthesia in patients with active bronchospasm. These reactions rarely occur with halothane and sevoflurane, which are considered nonpungent. Therefore, the bronchodilating action of halothane and sevoflurane makes them the agents of choice in patients with underlying airway problems. Nitrous oxide is also nonpungent and can facilitate inhalational induction of anesthesia in a patient with bronchospasm. The control of breathing is significantly affected by inhaled anesthetics. With the exception of nitrous oxide, all inhaled anesthetics in current use cause a dose-dependent decrease in tidal volume and an increase in respiratory rate, resulting in a rapid, shallow breathing pattern. However, the increase in respiratory rate varies among agents and does not fully compensate for the decrease in tidal volume, resulting in a decrease in alveolar ventilation. In addition, all volatile anesthetics are respiratory depressants, as defined by a reduced ventilatory response to increased levels of carbon dioxide in the blood. The degree of ventilatory depression varies among the volatile agents, with isoflurane and enflurane being the most depressant. By this hypoventilation mechanism, all volatile anesthetics increase the resting level of PaCO2 . Volatile anesthetics also raise the apneic threshold (Pa CO2 level below which apnea occurs through lack of CO2 -driven respiratory stimulation) and decrease the ventilatory response to hypoxia. In practice, the respiratory depressant effects of anesthetics are overcome by assisting (controlling) ventilation mechanically. The ventilatory depression produced by inhaled anesthetics may be counteracted by surgical stimulation; however, low, subanesthetic concentrations of volatile anesthetic present after surgery in the early recovery period can continue to depress the compensatory increase in ventilation normally caused by hypoxia. Inhaled anesthetics also depress mucociliary function in the airway. During prolonged exposure to inhaled anesthetics, mucus pooling and plugging may result in atelectasis and the development of postoperative respiratory complications, including hypoxemia and respiratory infections. D. Renal Effects Inhaled anesthetics tend to decrease glomerular filtration rate (GFR) and urine flow. Renal blood flow may also be decreased by some agents but filtration fraction is increased, implying that autoregulatory control of efferent arteriole tone helps compensate and limits the reduction in GFR. In general these anesthetic effects are minor compared with the stress of surgery itself and usually reversible after discontinuation of the anesthetic. E. Hepatic Effects Volatile anesthetics cause a concentration-dependent decrease in portal vein blood flow that parallels the decline in cardiac output produced by these agents. However, total hepatic blood flow may be relatively preserved as hepatic artery blood flow to the liver may increase or stay the same. Although transient changes in liver function tests may occur following exposure to volatile anesthetics, persistent elevation in liver enzymes is rare except following repeated exposures to halothane (see Toxicity of Anesthetic Agents). F. Effects on Uterine Smooth Muscle Nitrous oxide appears to have little effect on uterine musculature. However, the halogenated anesthetics are potent uterine muscle relaxants and produce this effect in a concentration-dependent fashion. This pharmacologic effect can be helpful when profound uterine relaxation is required for intrauterine fetal manipulation or manual extraction of a retained placenta during delivery. However, it can also lead to increased uterine bleeding.

Toxicity of Anesthetic Agents A. Acute Toxicity 1. Nephrotoxicity—Metabolism of enflurane and sevoflurane may generate compounds that are potentially nephrotoxic. Although their metabolism can liberate nephrotoxic fluoride ions, significant renal injury has been reported only for enflurane with prolonged exposure. The insolubility and rapid elimination of sevoflurane may prevent toxicity. This drug may be degraded by carbon dioxide absorbents in anesthesia machines to form a nephrotoxic vinyl ether compound termed “compound A” which, in high concentrations, has caused proximal tubular necrosis in rats. Nevertheless, there have been no reports of renal injury in humans receiving sevoflurane anesthesia. Moreover, exposure to sevoflurane does not produce any change in standard markers of renal function. 2. Hematotoxicity—Prolonged exposure to nitrous oxide decreases methionine synthase activity, which theoretically could cause megaloblastic anemia. Megaloblastic bone marrow changes have been observed in patients after 12-hour exposure to 50% nitrous oxide.


Chronic exposure of dental personnel to nitrous oxide in inadequately ventilated dental operating suites is a potential occupational hazard. All inhaled anesthetics can produce some carbon monoxide (CO) from their interaction with strong bases in dry carbon dioxide absorbers. CO binds to hemoglobin with high affinity, reducing oxygen delivery to tissues. Desflurane produces the most CO, and intraoperative formation of CO has been reported. CO production can be avoided simply by using fresh carbon dioxide absorbent and by preventing its complete desiccation. 3. Malignant hyperthermia—Malignant hyperthermia is a heritable genetic disorder of skeletal muscle that occurs in susceptible individuals exposed to volatile anesthetics while undergoing general anesthesia (see Chapter 16 and Table 16–4). The depolarizing muscle relaxant succinylcholine may also trigger malignant hyperthermia. The malignant hyperthermia syndrome consists of muscle rigidity, hyperthermia, rapid onset of tachycardia and hypercapnia, hyperkalemia, and metabolic acidosis following exposure to one or more triggering agents. Malignant hyperthermia is a rare but important cause of anesthetic morbidity and mortality. The specific biochemical abnormality is an increase in free cytosolic calcium concentration in skeletal muscle cells. Treatment includes administration of dantrolene (to reduce calcium release from the sarcoplasmic reticulum) and appropriate measures to reduce body temperature and restore electrolyte and acid-base balance (see Chapter 27). Malignant hyperthermia susceptibility is characterized by genetic heterogeneity, and several predisposing clinical myopathies have been identified. It has been associated with mutations in the gene coding for the skeletal muscle ryanodine receptor (RyR1, the calcium release channel on the sarcoplasmic reticulum), and mutant alleles of the gene encoding the α1 subunit of the human skeletal muscle Ltype voltage-dependent calcium channel. However, the genetic loci identified to date account for less than 50% of malignant hyperthermia-susceptible individuals, and genetic testing cannot definitively determine malignant hyperthermia susceptibility. Currently, the most reliable test to establish susceptibility is the in vitro caffeine-halothane contracture test using skeletal muscle biopsy samples. 4. Hepatotoxicity (halothane hepatitis)—Hepatic dysfunction following surgery and general anesthesia is most likely caused by hypovolemic shock, infection conferred by blood transfusion, or other surgical stresses rather than by volatile anesthetic toxicity. However, a small subset of individuals previously exposed to halothane has developed fulminant hepatic failure. The incidence of severe hepatotoxicity following exposure to halothane is estimated to be in the range of 1 in 20,000–35,000. The mechanisms underlying halothane hepatotoxicity remain unclear, but studies in animals implicate the formation of reactive metabolites that either cause direct hepatocellular damage (eg, free radicals) or initiate immune-mediated responses. Cases of hepatitis following exposure to other volatile anesthetics, including enflurane, isoflurane, and desflurane, have rarely been reported. B. Chronic Toxicity 1. Mutagenicity, teratogenicity, and reproductive effects —Under normal conditions, inhaled anesthetics including nitrous oxide are neither mutagens nor carcinogens in patients. Nitrous oxide can be directly teratogenic in animals under conditions of extremely high exposure. Halothane, enflurane, isoflurane, desflurane, and sevoflurane may be teratogenic in rodents as a result of physiologic changes associated with the anesthesia rather than through a direct teratogenic effect. The most consistent finding in surveys conducted to determine the reproductive success of female operating room personnel has been a questionably higher-than-expected incidence of miscarriages. However, there are several problems in interpreting these studies. The association of obstetric problems with surgery and anesthesia in pregnant patients is also an important consideration. In the USA, at least 50,000 pregnant women each year undergo anesthesia and surgery for indications unrelated to pregnancy. The risk of abortion is clearly higher following this experience. It is not obvious, however, whether the underlying disease, surgery, anesthesia, or a combination of these factors is the cause of the increased risk 2. Carcinogenicity—Epidemiologic studies suggested an increase in the cancer rate in operating room personnel who were exposed to trace concentrations of anesthetic agents. However, no study has demonstrated the existence of a causal relationship between anesthetics and cancer. Many other factors might account for the questionably positive results seen after a careful review of epidemiologic data. Most operating rooms now use scavenging systems to remove trace concentrations of anesthetics released from anesthetic machines.

INTRAVENOUS ANESTHETICS Intravenous nonopioid anesthetics play an essential role in the practice of modern anesthesia. They are used to facilitate rapid induction of anesthesia and have replaced inhalation as the preferred method of anesthesia induction in most settings except for pediatric anesthesia. Intravenous agents are also commonly used to provide sedation during monitored anesthesia care and for patients in ICU settings. With the introduction of propofol, intravenous anesthesia also became a good option for the maintenance of anesthesia. However, similar to the inhaled agents, the currently available intravenous anesthetics are not ideal anesthetic drugs in the sense of producing all and only the five desired effects (unconsciousness, amnesia, analgesia, inhibition of autonomic reflexes, and skeletal muscle relaxation). Therefore, balanced anesthesia employing multiple drugs (inhaled anesthetics, sedative-hypnotics, opioids, neuromuscular blocking drugs) is generally used to minimize unwanted effects.


The intravenous anesthetics used for induction of general anesthesia are lipophilic and preferentially partition into highly perfused lipophilic tissues (brain, spinal cord), which accounts for their rapid onset of action. Regardless of the extent and speed of their metabolism, termination of the effect of a single bolus is determined by redistribution of the drug into less perfused and inactive tissues such as skeletal muscle and fat. Thus, all drugs used for induction of anesthesia have a similar duration of action when administered as a single bolus dose despite significant differences in their metabolism. Figure 25–6 shows the chemical structures of common clinically used intravenous anesthetics. Table 25–2 lists pharmacokinetic properties of these and other intravenous agents.

FIGURE 25–6 Chemical structures of some intravenous anesthetics. TABLE 25–2 Pharmacokinetic properties of intravenous anesthetics.


PROPOFOL In most countries, propofol is the most frequently administered drug for induction of anesthesia and has largely replaced barbiturates for this use. Because its pharmacokinetic profile allows for continuous infusions, propofol is also used during maintenance of anesthesia and is a common choice for sedation in the setting of monitored anesthesia care. Increasingly, propofol is also used for sedation in the ICU as well as conscious sedation and short-duration general anesthesia in locations outside the operating room (eg, interventional radiology suites, emergency department; see Box: Sedation & Monitored Anesthesia Care, earlier). Propofol (2,6-diisopropylphenol) is an alkyl phenol with hypnotic properties that is chemically distinct from other groups of intravenous anesthetics (Figure 25–6). Because of its poor solubility in water, it is formulated as an emulsion containing 10% soybean oil, 2.25% glycerol, and 1.2% lecithin, the major component of the egg yolk phosphatide fraction. Hence, susceptible patients may experience allergic reactions. The solution appears milky white and slightly viscous, has a pH of approximately 7, and a propofol concentration of 1% (10 mg/mL). In some countries, a 2% formulation is available. Although retardants of bacterial growth are added to the formulations, solutions should be used as soon as possible (no more than 8 hours after opening the vial) and proper sterile technique is essential. The addition of metabisulfite in one of the formulations has raised concern regarding its use in patients with reactive airway disease (eg, asthma) or sulfite allergies. The presumed mechanism of action of propofol is through potentiation of the chloride current mediated through the GABAA receptor complex.

Pharmacokinetics Propofol is rapidly metabolized in the liver; the resulting water-soluble compounds are presumed to be inactive and are excreted through the kidneys. Plasma clearance is high and exceeds hepatic blood flow, indicating the importance of extrahepatic metabolism, which presumably occurs in the lungs and may account for the elimination of up to 30% of a bolus dose of the drug (Table 25–2). The recovery from propofol is more complete, with less “hangover” than that observed with thiopental, likely due to the high plasma clearance.


However, as with other intravenous drugs, transfer of propofol from the plasma (central) compartment and the associated termination of drug effect after a single bolus dose are mainly the result of redistribution from highly perfused (brain) to less-well-perfused (skeletal muscle) compartments (Figure 25–7). As with other intravenous agents, awakening after an induction dose of propofol usually occurs within 8–10 minutes. The kinetics of propofol (and other intravenous anesthetics) after a single bolus dose or continuous infusion are best described by means of a three-compartment model. Such models have been used as the basis for developing systems of target-controlled infusions.

FIGURE 25–7 Redistribution of thiopental after an intravenous bolus administration. The redistribution curves for bolus administration of other intravenous anesthetics are similar, explaining the observation that recovery times are the same despite remarkable differences in metabolism. Note that the time axis is not linear. The context-sensitive half-time of a drug describes the elimination half-time after discontinuation of a continuous infusion as a function of the duration of the infusion. It is an important factor in the suitability of a drug for use as maintenance anesthetic. The context-sensitive half-time of propofol is brief, even after a prolonged infusion, and therefore recovery occurs relatively promptly (Figure 25–8).


FIGURE 25–8 The context-sensitive half-time of common intravenous anesthetics. Even after a prolonged infusion, the half-time of propofol is relatively short, which makes propofol the preferred choice for intravenous anesthesia. Ketamine and etomidate have similar characteristics but their use is limited by other effects.

Organ System Effects A. CNS Effects Propofol acts as hypnotic but does not have analgesic properties. Although the drug leads to a general suppression of CNS activity, excitatory effects such as twitching or spontaneous movement are occasionally observed during induction of anesthesia. These effects may resemble seizure activity; however, most studies support an anticonvulsant effect of propofol, and the drug may be safely administered to patients with seizure disorders. Propofol decreases cerebral blood flow and the cerebral metabolic rate for oxygen (CMRO2 ), which decreases intracranial pressure (ICP) and intraocular pressure; the magnitude of these changes is comparable to that of thiopental. Although propofol can produce a desired decrease in ICP, the combination of reduced cerebral blood flow and reduced mean arterial pressure due to peripheral vasodilation can critically decrease cerebral perfusion pressure. When administered in large doses, propofol produces burst suppression in the EEG, an end point that has been used when administering intravenous anesthetics for neuroprotection during neurosurgical procedures. Evidence from animal studies suggests that propofol’s neuroprotective effects during focal ischemia are similar to those of thiopental and isoflurane. B. Cardiovascular Effects Compared with other induction drugs, propofol produces the most pronounced decrease in systemic blood pressure; this is a result of profound vasodilation in both arterial and venous circulations leading to reductions in preload and afterload. This effect on systemic blood pressure is more pronounced with increased age, in patients with reduced intravascular fluid volume, and with rapid injection. Because the hypotensive effects are further augmented by the inhibition of the normal baroreflex response, the vasodilation only leads to a small increase in heart rate. In fact, profound bradycardia and asystole after the administration of propofol have been described in healthy adults despite prophylactic anticholinergic drugs. C. Respiratory Effects Propofol is a potent respiratory depressant and generally produces apnea after an induction dose. A maintenance infusion reduces minute ventilation through reductions in tidal volume and respiratory rate, with the effect on tidal volume being more pronounced. In addition, the ventilatory response to hypoxia and hypercapnia is reduced. Propofol causes a greater reduction in upper airway reflexes than thiopental does, which makes it well suited for instrumentation of the airway, such as placement of a laryngeal mask airway. D. Other Effects Although propofol, unlike volatile anesthetics, does not augment neuromuscular block, studies have found good intubating conditions after propofol induction without the use of neuromuscular blocking agents. Unexpected tachycardia occurring during propofol anesthesia


should prompt laboratory evaluation for possible metabolic acidosis (propofol infusion syndrome). An interesting and desirable side effect of propofol is its antiemetic activity. Pain on injection is a common complaint and can be reduced by premedication with an opioid or coadministration with lidocaine. Dilution of propofol and the use of larger veins for injection can also reduce the incidence and severity of injection pain.

Clinical Uses & Dosage The most common use of propofol is to facilitate induction of general anesthesia by bolus injection of 1–2.5 mg/kg IV. Increasing age, reduced cardiovascular reserve, or premedication with benzodiazepines or opioids reduces the required induction dose; children require higher doses (2.5–3.5 mg/kg IV). Generally, titration of the induction dose helps to prevent severe hemodynamic changes. Propofol is often used for maintenance of anesthesia either as part of a balanced anesthesia regimen in combination with volatile anesthetics, nitrous oxide, sedative-hypnotics, and opioids or as part of a total intravenous anesthetic technique, usually in combination with opioids. Therapeutic plasma concentrations for maintenance of anesthesia normally range between 3 and 8 mcg/mL (typically requiring a continuous infusion rate between 100 and 200 mcg/kg/min) when combined with nitrous oxide or opioids. When used for sedation of mechanically ventilated patients in the ICU or for sedation during procedures, the required plasma concentration is 1–2 mcg/mL, which can be achieved with a continuous infusion at 25–75 mcg/kg/min. Because of its pronounced respiratory depressant effect and narrow therapeutic range, propofol should be administered only by individuals trained in airway management. Subanesthetic doses of propofol can be used to treat postoperative nausea and vomiting (10–20 mg IV as bolus or 10 mcg/kg/min as an infusion).

FOSPROPOFOL As previously noted, injection pain during administration of propofol is often perceived as severe, and the lipid emulsion has several disadvantages. Intense research has focused on finding alternative formulations or related drugs that would address some of these problems. Fospropofol is a water-soluble prodrug of propofol, rapidly metabolized by alkaline phosphatase, and producing propofol, phosphate, and formaldehyde. The formaldehyde is metabolized by aldehyde dehydrogenase in the liver and in erythrocytes. The available fospropofol formulation is a sterile, aqueous, colorless, and clear solution that is supplied in a single-dose vial at a concentration of 35 mg/mL under the trade name Lusedra.

Pharmacokinetics & Organ System Effects Because the active compound is propofol and fospropofol is a prodrug that requires metabolism to form propofol, the pharmacokinetics are more complex than for propofol itself. Multi-compartment models with two compartments for fospropofol and three for propofol have been used to describe the kinetics. The effect profile is similar to that of propofol, but onset and recovery are prolonged compared with propofol because the prodrug must first be converted into an active form. Although patients receiving fospropofol do not appear to experience the injection pain typical of propofol, a common adverse effect is the experience of paresthesia, often in the perianal region, which occurs in up to 74% of patients. The mechanism for this effect is unknown.

Clinical Uses & Dosage Fospropofol is approved for sedation during monitored anesthesia care. Supplemental oxygen must be administered to all patients receiving the drug. As with propofol, airway compromise is a major concern. Hence, it is recommended that fospropofol be administered only by personnel trained in airway management. The recommended standard dosage is an initial bolus dose of 6.5 mg/kg IV followed by supplemental doses of 1.6 mg/kg IV as needed. For patients weighing more than 90 kg or less than 60 kg, 90 or 60 kg should be used to calculate the dose, respectively. The dose should be reduced by 25% in patients older than 65 years and in those with an American Society of Anesthesiologists status of 3 or 4.

BARBITURATES This section focuses on the use of thiopental and methohexital for induction of general anesthesia; however, these barbiturate hypnotics have been largely replaced as induction agents by propofol. Other barbiturates and general barbiturate pharmacology are discussed in Chapter 22. The anesthetic effect of barbiturates presumably involves a combination of enhancement of inhibitory and inhibition of excitatory neurotransmission (Figure 25–1). Although the effects on inhibitory transmission probably result from activation of the GABA A receptor


complex, the effects on excitatory transmission are less well understood.

Pharmacokinetics Thiopental and methohexital undergo hepatic metabolism, mostly by oxidation but also by N-dealkylation, desulfuration, and destruction of the barbituric acid ring structure. Barbiturates should not be administered to patients with acute intermittent porphyria because they increase the production of porphyrins through stimulation of aminolevulinic acid synthetase. Methohexital has a shorter elimination halftime than thiopental due to its larger plasma clearance (Table 25–2), leading to a faster and more complete recovery after bolus injection. Although thiopental is metabolized more slowly and has a long elimination half-time, recovery after a single bolus injection is comparable to that of methohexital and propofol because it depends on redistribution to inactive tissue sites rather than on metabolism (Figure 25–7). However, if administered through repeated bolus injections or continuous infusion, recovery will be markedly prolonged because elimination will depend on metabolism under these circumstances (see also context-sensitive half-time, Figure 25–8).

Organ System Effects A. CNS Effects Barbiturates produce dose-dependent CNS depression ranging from sedation to general anesthesia when administered as bolus injections. They do not produce analgesia; instead, some evidence suggests they may reduce the pain threshold causing hyperalgesia. Barbiturates are potent cerebral vasoconstrictors and produce predictable decreases in cerebral blood flow, cerebral blood volume, and ICP. As a result, they decrease CMRO2 consumption in a dose-dependent manner up to a dose at which they suppress all EEG activity. The ability of barbiturates to decrease ICP and CMRO 2 makes these drugs useful in the management of patients with space-occupying intracranial lesions. They may provide neuroprotection from focal cerebral ischemia (stroke, surgical retraction, temporary clips during aneurysm surgery), but probably not from global cerebral ischemia (eg, from cardiac arrest). Except for methohexital, barbiturates decrease electrical activity on the EEG and can be used as anticonvulsants. In contrast, methohexital activates epileptic foci and may therefore be useful to facilitate electroconvulsive therapy or during the identification of epileptic foci during surgery. B. Cardiovascular Effects The decrease in systemic blood pressure associated with administration of barbiturates for induction of anesthesia is primarily due to peripheral vasodilation and is usually smaller than the blood pressure decrease associated with propofol. There are also direct negative inotropic effects on the heart. However, inhibition of the baroreceptor reflex is less pronounced than with propofol; thus, compensatory increases in heart rate limit the decrease in blood pressure and make it transient. The depressant effects on systemic blood pressure are increased in patients with hypovolemia, cardiac tamponade, cardiomyopathy, coronary artery disease, or cardiac valvular disease because such patients are less able to compensate for the effects of peripheral vasodilation. Hemodynamic effects are also more pronounced with larger doses and rapid injection. C. Respiratory Effects Barbiturates are respiratory depressants, and a usual induction dose of thiopental or methohexital typically produces transient apnea, which will be more pronounced if other respiratory depressants are also administered. Barbiturates lead to decreased minute ventilation through reduced tidal volumes and respiratory rate and also decrease the ventilatory responses to hypercapnia and hypoxia. Resumption of spontaneous breathing after an anesthetic induction dose of a barbiturate is characterized by a slow breathing rate and decreased tidal volume. Suppression of laryngeal reflexes and cough reflexes is probably not as profound as after an equianesthetic propofol administration, which makes barbiturates an inferior choice for airway instrumentation in the absence of neuromuscular blocking drugs. Furthermore, stimulation of the upper airway or trachea (eg, by secretions, laryngeal mask airway, direct laryngoscopy, tracheal intubation) during inadequate depression of airway reflexes may result in laryngospasm or bronchospasm. This phenomenon is not unique to barbiturates but is true whenever the drug dose is inadequate to suppress the airway reflexes. D. Other Effects Accidental intra-arterial injection of barbiturates results in excruciating pain and intense vasoconstriction, often leading to severe tissue injury involving gangrene. Approaches to treatment include blockade of the sympathetic nervous system (eg, stellate ganglion block) in the involved extremity. If extravasation occurs, some authorities recommend local injection of the area with 0.5% lidocaine (5–10 mL) in an attempt to dilute the barbiturate concentration. Life-threatening allergic reactions to barbiturates are rare, with an estimated occurrence of 1 in 30,000 patients. However, barbiturate-induced histamine release occasionally is seen.

Clinical Uses & Dosage The principal clinical use of thiopental (3–5 mg/kg IV) or methohexital (1–1.5 mg/kg IV) is for induction of anesthesia (unconsciousness),


which usually occurs in less than 30 seconds. Patients may experience a garlic or onion taste after administration. Solutions of thiopental sodium for intravenous injection have a pH range of 10–11 to maintain stability. Rapid co-injection with depolarizing and nondepolarizing muscle relaxants, which have much lower pH, may cause precipitation of insoluble thiopentone acid. Barbiturates such as methohexital (20–30 mg/kg) may be administered per rectum to facilitate induction of anesthesia in mentally challenged and uncooperative pediatric patients. When a barbiturate is administered with the goal of neuroprotection, an isoelectric EEG indicating maximal reduction of CMRO2 has traditionally been used as the end point. More recent data demonstrating equal protection after smaller doses have challenged this practice. The use of smaller doses is less frequently associated with hypotension, thus making it easier to maintain adequate cerebral perfusion pressure, especially in the setting of increased ICP.

BENZODIAZEPINES Benzodiazepines commonly used in the perioperative period include midazolam, lorazepam, and less frequently, diazepam. Benzodiazepines are unique among the group of intravenous anesthetics in that their action can readily be terminated by administration of their selective antagonist, flumazenil. Their most desired effects are anxiolysis and anterograde amnesia, which are extremely useful for premedication. The chemical structure and pharmacodynamics of the benzodiazepines are discussed in detail in Chapter 22.

Pharmacokinetics in the Anesthesia Setting The highly lipid-soluble benzodiazepines rapidly enter the CNS, which accounts for their rapid onset of action, followed by redistribution to inactive tissue sites and subsequent termination of the drug effect. Additional information regarding the pharmacokinetics of the benzodiazepines may be found in Chapter 22. Despite its prompt passage into the brain, midazolam is considered to have a slower effect-site equilibration time than propofol and thiopental. In this regard, intravenous doses of midazolam should be sufficiently spaced to permit the peak clinical effect to be recognized before a repeat dose is considered. Midazolam has the shortest context-sensitive half-time, which makes it the only one of the three benzodiazepine drugs suitable for continuous infusion (Figure 25–8).

Organ System Effects A. CNS Effects Similar to propofol and barbiturates, benzodiazepines decrease CMRO2 and cerebral blood flow, but to a smaller extent. There appears to be a ceiling effect for benzodiazepine-induced decreases in CMRO2 as evidenced by midazolam’s inability to produce an isoelectric EEG. Patients with decreased intracranial compliance demonstrate little or no change in ICP after the administration of midazolam. Although neuroprotective properties have not been shown for benzodiazepines, these drugs are potent anticonvulsants used in the treatment of status epilepticus, alcohol withdrawal, and local anesthetic-induced seizures. The CNS effects of benzodiazepines can be promptly terminated by administration of the selective benzodiazepine antagonist flumazenil, which improves their safety profile. B. Cardiovascular Effects If used for the induction of anesthesia, midazolam produces a greater decrease in systemic blood pressure than comparable doses of diazepam. These changes are most likely due to peripheral vasodilation inasmuch as cardiac output is not changed. Similar to other intravenous induction agents, midazolam’s effect on systemic blood pressure is exaggerated in hypovolemic patients. C. Respiratory Effects Benzodiazepines produce minimal depression of ventilation, although transient apnea may follow rapid intravenous administration of midazolam for induction of anesthesia, especially in the presence of opioid premedication. Benzodiazepines decrease the ventilatory response to carbon dioxide, but this effect is not usually significant if they are administered alone. More severe respiratory depression can occur when benzodiazepines are administered together with opioids. Another problem affecting ventilation is airway obstruction induced by the hypnotic effects of benzodiazepines. D. Other Effects Pain during intravenous and intramuscular injection and subsequent thrombophlebitis are most pronounced with diazepam and reflect the poor water solubility of this benzodiazepine, which requires an organic solvent in the formulation. Despite its better solubility (which eliminates the need for an organic solvent), midazolam may also produce pain on injection. Allergic reactions to benzodiazepines are rare to nonexistent.


Clinical Uses & Dosage Benzodiazepines are most commonly used for preoperative medication, intravenous sedation, and suppression of seizure activity. Less frequently, midazolam and diazepam may also be used to induce general anesthesia. The slow onset and prolonged duration of action of lorazepam limit its usefulness for preoperative medication or induction of anesthesia, especially when rapid and sustained awakening at the end of surgery is desirable. Although flumazenil (8–15 mcg/kg IV) may be useful for treating patients experiencing delayed awakening, its duration of action is brief (about 20 minutes) and resedation may occur. The amnestic, anxiolytic, and sedative effects of benzodiazepines make this class of drugs the most popular choice for preoperative medication. Midazolam (1–2 mg IV) is effective for premedication, sedation during regional anesthesia, and brief therapeutic procedures. Midazolam has a more rapid onset, with greater amnesia and less postoperative sedation, than diazepam. Midazolam is also the most commonly used oral premedication for children; 0.5 mg/kg administered orally 30 minutes before induction of anesthesia provides reliable sedation and anxiolysis in children without producing delayed awakening. The synergistic effects between benzodiazepines and other drugs, especially opioids and propofol, can be used to achieve better sedation and analgesia but may also greatly enhance their combined respiratory depression and may lead to airway obstruction or apnea. Because benzodiazepine effects are more pronounced with increasing age, dose reduction and careful titration may be necessary in elderly patients. General anesthesia can be induced by the administration of midazolam (0.1–0.3 mg/kg IV), but the onset of unconsciousness is slower than after the administration of thiopental, propofol, or etomidate. Delayed awakening is a potential disadvantage, limiting the usefulness of benzodiazepines for induction of general anesthesia despite their advantage of less pronounced circulatory effects.

ETOMIDATE Etomidate (Figure 25–6) is an intravenous anesthetic with hypnotic but not analgesic effects and is often chosen for its minimal hemodynamic effects. Although its pharmacokinetics are favorable, endocrine side effects limit its use for continuous infusions. Etomidate is a carboxylated imidazole derivative that is poorly soluble in water and is therefore supplied as a 2 mg/mL solution in 35% propylene glycol. The solution has a pH of 6.9 and does not cause problems with precipitation as thiopental does. Etomidate appears to have GABA-like effects and seems to act primarily through potentiation of GABAA-mediated chloride currents, like most other intravenous anesthetics.

Pharmacokinetics An induction dose of etomidate produces rapid onset of anesthesia, and recovery depends on redistribution to inactive tissue sites, comparable to thiopental and propofol. Metabolism is primarily by ester hydrolysis to inactive metabolites, which are then excreted in urine (78%) and bile (22%). Less than 3% of an administered dose of etomidate is excreted as unchanged drug in urine. Clearance of etomidate is about five times that of thiopental, as reflected by a shorter elimination half-time (Table 25–2). The duration of action is linearly related to the dose, with each 0.1 mg/kg providing about 100 seconds of unconsciousness. Because of etomidate’s minimal effects on hemodynamics and short context-sensitive half-time, larger doses, repeated boluses, or continuous infusions can safely be administered. Etomidate, like most other intravenous anesthetics, is highly protein bound (77%), primarily to albumin.

Organ System Effects A. CNS Effects Etomidate is a potent cerebral vasoconstrictor, as reflected by decreases in cerebral blood flow and ICP. These effects are similar to those produced by comparable doses of thiopental. Despite its reduction of CMRO2 , etomidate has failed to show neuroprotective properties in animal studies, and human studies are lacking. The frequency of excitatory spikes on the EEG after the administration of etomidate is greater than with thiopental. Similar to methohexital, etomidate may activate seizure foci, manifested as fast activity on the EEG. In addition, spontaneous movements characterized as myoclonus occur in more than 50% of patients receiving etomidate, and this myoclonic activity may be associated with seizure-like activity on the EEG. B. Cardiovascular Effects A characteristic and desired feature of induction of anesthesia with etomidate is cardiovascular stability after bolus injection. In this regard, decrease in systemic blood pressure is modest or absent and principally reflects a decrease in systemic vascular resistance. Therefore, the systemic blood pressure-lowering effects of etomidate are probably exaggerated in the presence of hypovolemia, and the patient’s intravascular fluid volume status should be optimized before induction of anesthesia. Etomidate produces minimal changes in heart rate and cardiac output. Its depressant effects on myocardial contractility are minimal at concentrations used for induction of anesthesia.


C. Respiratory Effects The depressant effects of etomidate on ventilation are less pronounced than those of barbiturates, although apnea may occasionally follow rapid intravenous injection of the drug. Depression of ventilation may be exaggerated when etomidate is combined with inhaled anesthetics or opioids. D. Endocrine Effects Etomidate causes adrenocortical suppression by producing a dose-dependent inhibition of 11β-hydroxylase, an enzyme necessary for the conversion of cholesterol to cortisol (see Figure 39–1). This suppression lasts 4–8 hours after an induction dose of the drug. Despite concerns regarding this finding, no outcome studies have demonstrated an adverse effect when etomidate is given in a bolus dose. However, because of its endocrine effects, etomidate is not used as continuous infusion.

Clinical Uses & Dosage Etomidate is an alternative to propofol and barbiturates for the rapid intravenous induction of anesthesia, especially in patients with compromised myocardial contractility. After a standard induction dose (0.2–0.3 mg/kg IV), the onset of unconsciousness is comparable to that achieved by thiopental and propofol. Similar to propofol, during intravenous injection of etomidate there is a high incidence of pain, which may be followed by venous irritation. Involuntary myoclonic movements are also common but may be masked by the concomitant administration of neuromuscular blocking drugs. Awakening after a single intravenous dose of etomidate is rapid, with little evidence of any residual depressant effects. Etomidate does not produce analgesia, and postoperative nausea and vomiting may be more common than after the administration of thiopental or propofol.

KETAMINE Ketamine (Figure 25–6) is a partially water-soluble and highly lipid-soluble phencyclidine derivative differing from most other intravenous anesthetics in that it produces significant analgesia. The characteristic state observed after an induction dose of ketamine is known as “dissociative anesthesia,” wherein the patient’s eyes remain open with a slow nystagmic gaze (cataleptic state). Of the two stereoisomers, the S(+) form is more potent than the R(−) isomer, but only the racemic mixture of ketamine is available in the USA. Ketamine’s mechanism of action is complex, but the major effect is probably produced through inhibition of the NMDA receptor complex.

Pharmacokinetics The high lipid solubility of ketamine ensures a rapid onset of its effect. As with other intravenous induction drugs, the effect of a single bolus injection is terminated by redistribution to inactive tissue sites. Metabolism occurs primarily in the liver and involves Ndemethylation by the cytochrome P450 system. Norketamine, the primary active metabolite, is less potent (one third to one fifth the potency of ketamine) and is subsequently hydroxylated and conjugated into water-soluble inactive metabolites that are excreted in urine. Ketamine is the only intravenous anesthetic that has low protein binding (Table 25–2).

Organ System Effects If ketamine is administered as the sole anesthetic, amnesia is not as complete as with the benzodiazepines. Reflexes are often preserved, but it cannot be assumed that patients are able to protect the upper airway. The eyes remain open and the pupils are moderately dilated with a nystagmic gaze. Frequently, lacrimation and salivation are increased, and premedication with an anticholinergic drug may be indicated to limit this effect. A. CNS Effects In contrast to other intravenous anesthetics, ketamine is considered to be a cerebral vasodilator that increases cerebral blood flow, as well as CMRO2 . For these reasons, ketamine has traditionally not been recommended for use in patients with intracranial pathology, especially increased ICP. Nevertheless, these perceived undesirable effects on cerebral blood flow may be blunted by the maintenance of normocapnia. Despite the potential to produce myoclonic activity, ketamine is considered an anticonvulsant and may be recommended for treatment of status epilepticus when more conventional drugs are ineffective. Unpleasant emergence reactions after administration are the main factor limiting ketamine’s use. Such reactions may include vivid colorful dreams, hallucinations, out-of-body experiences, and increased and distorted visual, tactile, and auditory sensitivity. These reactions can be associated with fear and confusion, but a euphoric state may also be induced, which explains the potential for abuse of the drug. Children usually have a lower incidence of and less severe emergence reactions. Combination with a benzodiazepine may be indicated to limit the unpleasant emergence reactions and also increase amnesia.


B. Cardiovascular Effects Ketamine can produce transient but significant increases in systemic blood pressure, heart rate, and cardiac output, presumably by centrally mediated sympathetic stimulation. These effects, which are associated with increased cardiac workload and myocardial oxygen consumption, are not always desirable and can be blunted by coadministration of benzodiazepines, opioids, or inhaled anesthetics. Though the effect is more controversial, ketamine is also considered to be a direct myocardial depressant. This property is usually masked by its stimulation of the sympathetic nervous system but may become apparent in critically ill patients with limited ability to increase their sympathetic nervous system activity. C. Respiratory Effects Ketamine is not thought to produce significant respiratory depression. When it is used as a single drug, the respiratory response to hypercapnia is preserved and blood gases remain stable. Transient hypoventilation and, in rare cases, a short period of apnea can follow rapid administration of a large intravenous dose for induction of anesthesia. The ability to protect the upper airway in the presence of ketamine cannot be assumed despite the presence of active airway reflexes. Especially in children, the risk for laryngospasm because of increased salivation must be considered; this risk can be reduced by premedication with an anticholinergic drug. Ketamine relaxes bronchial smooth muscles and may be helpful in patients with reactive airways and in the management of patients experiencing bronchoconstriction.

Clinical Uses & Dosage Its unique properties, including profound analgesia, stimulation of the sympathetic nervous system, bronchodilation, and minimal respiratory depression, make ketamine an important alternative to the other intravenous anesthetics and a desirable adjunct in many cases despite the unpleasent psychotomimetic effects. Moreover, ketamine can be administered by multiple routes (intravenous, intramuscular, oral, rectal, epidural), thus making it a useful option for premedication in mentally challenged and uncooperative pediatric patients. Induction of anesthesia can be achieved with ketamine, 1–2 mg/kg intravenously or 4–6 mg/kg intramuscularly. Though the drug is not commonly used for maintenance of anesthesia, its short context-sensitive half-time makes ketamine a candidate for this purpose. For example, general anesthesia can be achieved with the infusion of ketamine, 15–45 mcg/kg/min, plus 50–70% nitrous oxide or by ketamine alone, 30–90 mcg/kg/min. Small bolus doses of ketamine (0.2–0.8 mg/kg IV) may be useful during regional anesthesia when additional analgesia is needed (eg, cesarean delivery under neuraxial anesthesia with an insufficient regional block). Ketamine provides effective analgesia without compromise of the airway. An infusion of a subanalgesic dose of ketamine (3–5 mcg/kg/min) during general anesthesia and in the early postoperative period may be useful to produce analgesia or reduce opioid tolerance and opioid-induced hyperalgesia. The use of ketamine has always been limited by its unpleasant psychotomimetic side effects, but its unique features make it a very valuable alternative in certain settings, mostly because of the potent analgesia with minimal respiratory depression. Most recently it has become popular as an adjunct administered at subanalgesic doses to limit or reverse opioid tolerance.

DEXMEDETOMIDINE Dexmedetomidine is a highly selective α2 -adrenergic agonist. Recognition of the usefulness of α2 agonists is based on observations of decreased anesthetic requirements in patients receiving chronic clonidine therapy. The effects of dexmedetomidine can be antagonized with α2 -antagonist drugs. Dexmedetomidine is the active S-enantiomer of medetomidine, a highly selective α2 -adrenergic agonist imidazole derivative that is used in veterinary medicine. Dexmedetomidine is water soluble and available as a parenteral formulation.

Pharmacokinetics Dexmedetomidine undergoes rapid hepatic metabolism involving N-methylation and hydroxylation, followed by conjugation. Metabolites are excreted in the urine and bile. Clearance is high, and the elimination half-time is short (Table 25–2). However, there is a significant increase in the context-sensitive half-time from 4 minutes after a 10-minute infusion to 250 minutes after an 8-hour infusion.

Organ System Effects A. CNS Effects Dexmedetomidine produces its selective α2 -agonist effects through activation of CNS α2 receptors. Hypnosis presumably results from stimulation of α2 receptors in the locus caeruleus, and the analgesic effect originates at the level of the spinal cord. The sedative effect produced by dexmedetomidine has a different quality than that produced by other intravenous anesthetics in that it more completely resembles a physiologic sleep state through activation of endogenous sleep pathways. Dexmedetomidine is likely to be associated with a


decrease in cerebral blood flow without significant changes in ICP and CMRO 2 . It has the potential to lead to the development of tolerance and dependence. B. Cardiovascular Effects Dexmedetomidine infusion results in moderate decreases in heart rate and systemic vascular resistance and, consequently, a decrease in systemic blood pressure. A bolus injection may produce a transient increase in systemic blood pressure and pronounced decrease in heart rate, an effect that is probably mediated through activation of peripheral α2 adrenoceptors. Bradycardia associated with dexmedetomidine infusion may require treatment. Heart block, severe bradycardia, and asystole have been observed and may result from unopposed vagal stimulation. The response to anticholinergic drugs is unchanged. C. Respiratory Effects The effects of dexmedetomidine on the respiratory system are a small to moderate decrease in tidal volume and very little change in the respiratory rate. The ventilatory response to carbon dioxide is unchanged. Although the respiratory effects are mild, upper airway obstruction as a result of sedation is possible. In addition, dexmedetomidine has a synergistic sedative effect when combined with other sedative-hypnotics.

Clinical Uses & Dosage Dexmedetomidine is principally used for the short-term sedation of intubated and ventilated patients in an ICU setting. In the operating room, dexmedetomidine may be used as an adjunct to general anesthesia or to provide sedation, eg, during awake fiberoptic tracheal intubation or regional anesthesia. When administered during general anesthesia, dexmedetomidine (0.5–1 mcg/kg loading dose over 10– 15 minutes, followed by an infusion of 0.2–0.7 mcg/kg/h) decreases the dose requirements for inhaled and injected anesthetics. Awakening and the transition to the postoperative setting may benefit from dexmedetomidine-produced sedative and analgesic effects without respiratory depression.

PREPARATIONS AVAILABLE*


OPIOID ANALGESICS Opioids are analgesic agents and are distinct from general anesthetics and hypnotics. Even when high doses of opioid analgesics are administered, recall cannot be prevented reliably unless hypnotic agents such as benzodiazepines are also used. Opioid analgesics are routinely used to achieve postoperative analgesia and intraoperatively as part of a balanced anesthesia regimen as described earlier (see Intravenous Anesthetics). Their pharmacology and clinical use are described in greater detail in Chapter 31. In addition to their use as part of a balanced anesthesia regimen, opioids in large doses have been used in combination with large doses of benzodiazepines to achieve a general anesthetic state, particularly in patients with limited circulatory reserve who undergo cardiac surgery. When administered in large doses, potent opioids such as fentanyl can induce chest wall (and laryngeal) rigidity, thereby acutely impairing mechanical ventilation. Furthermore, large doses of potent opioids may speed up the development of tolerance and complicate postoperative pain management.

CURRENT CLINICAL PRACTICE


The practice of clinical anesthesia requires integrating the pharmacology and the known adverse effects of these potent drugs with the pathophysiologic state of individual patients. Every case tests the ability of the anesthesiologist to produce the depth of anesthesia required to allow invasive surgery to proceed, despite major medical problems.

REFERENCES Allaert SE et al: First trimester anesthesia exposure and fetal outcome. A review. Acta Anaesthesiol Belg 2007;58:119. Ebert T J et al: Desflurane-mediated sympathetic activation occurs in humans despite preventing hypotension and baroreceptor unloading. Anesthesiology 1998;85:1227. Eger EI II: Uptake and distribution. In: Miller RD (editor): Anesthesia, 7th ed. Churchill Livingstone, 2010. Eger EI II, Saidman LJ, Brandstater B: Minimum alveolar anesthetic concentration: A standard of anesthetic potency. Anesthesiology 1965;26:756. Fraga M et al: T he effects of isoflurane and desflurane on intracranial pressure, cerebral perfusion and cerebral arteriovenous oxygen content difference in normocapnic patients with supratentorial brain tumors. Anesthesiology 2003;98:1085. Fragen RJ: Drug Infusions in Anesthesiology. Lippincott Williams & Wilkins, 2005. Hemmings HC et al: Emerging molecular mechanisms of general anesthetic action. T rends Pharmacol Sci 2005;26:503. Hirshey Dirksen SJ et al: Future directions in malignant hyperthermia research and patient care. Anesth Analg 2011;113:1108. Lugli AK, Yost CS, Kindler CH: Anesthetic mechanisms: Update on the challenge of unravelling the mystery of anaesthesia. Eur J Anaesth 2009;26:807. Olkkola KT , Ahonen J: Midazolam and other benzodiazepines. Handb Exp Pharmacol 2008;182:335. Reves JG et al: Intravenous anesthetics. In: Miller RD (editor): Anesthesia, 7th ed. Churchill Livingstone, 2010. Rudolph U et al: Sedatives, anxiolytics, and amnestics. In: Evers AS, Maze M (editors): Anesthetic Pharmacology: Physiologic Principles and Clinical Practice. Churchill Livingstone, 2004. Sjogren D, Lindahl SGE, Sollevi A: Ventilatory responses to acute and sustained hypoxia during isoflurane anesthesia. Anesth Analg 1998;86:403. Stoelting R, Hillier S: Barbiturates. In: Stoelting RK, Hillier SC (editors): Pharmacology and Physiology in Anesthetic Practice. Lippincott Williams & Wilkins, 2005. Yasuda N et al: Kinetics of desflurane, isoflurane, and halothane in humans. Anesthesiology 1991;70:489.

CASE STUDY ANSWER This patient presents with significant underlying cardiac risk and is scheduled to undergo major stressful surgery. Balanced anesthesia would begin with intravenous agents that cause minimal changes in blood pressure and heart rate such as a lowered dose of propofol or etomidate, combined with potent analgesics such as fentanyl (see Chapter 31) to block undesirable stimulation of autonomic reflexes. Maintenance of anesthesia could incorporate inhaled anesthetics that ensure unconsciousness and amnesia, additional intravenous agents to provide intraoperative and postoperative analgesia, and, if needed, neuromuscular blocking drugs (see Chapter 27) to induce muscle relaxation. The choice of inhaled agent(s) would be made based on the desire to maintain sufficient myocardial contractility, systemic blood pressure, and cardiac output for adequate perfusion of critical organs throughout the operation. If the patient’s ischemic pain has been chronic and severe, a low-dose ketamine infusion may be administered for additional pain control. Rapid emergence from the combined effects of the chosen anesthetic drugs, which would facilitate the patient’s return to a baseline state of heart function, breathing, and mentation, can be attained by understanding the known pharmacokinetic properties of the anesthetic agents as presented in this chapter.


CHAPTER

26 Local Anesthetics Kenneth Drasner, MD*

CASE STUDY A 67-year-old woman is scheduled for elective total knee arthroplasty. What local anesthetic agents would be most appropriate if surgical anesthesia were to be administered using a spinal or an epidural technique, and what potential complications might arise from their use? What anesthetics would be most appropriate for providing postoperative analgesia via an indwelling epidural or peripheral nerve catheter?

Simply stated, local anesthesia refers to loss of sensation in a limited region of the body. This is accomplished by disruption of afferent neural traffic via inhibition of impulse generation or propagation. Such blockade may bring with it other physiologic changes such as muscle paralysis and suppression of somatic or visceral reflexes, and these effects might be desirable or undesirable depending on the particular circumstances. Nonetheless, in most cases, it is the loss of sensation, or at least the achievement of localized analgesia, that is the primary goal. Although local anesthetics are often used as analgesics, it is their ability to provide complete loss of all sensory modalities that is their distinguishing characteristic. The contrast with general anesthesia should be obvious, but it is perhaps worthwhile to emphasize that with local anesthesia the drug is delivered directly to the target organ, and the systemic circulation serves only to diminish or terminate its effect. Local anesthesia can also be produced by various chemical or physical means. However, in routine clinical practice, it is achieved with a rather narrow spectrum of compounds, and recovery is normally spontaneous, predictable, and without residual effects. The development of these compounds has a rich history (see Box: Historical Development of Local Anesthesia), punctuated by serendipitous observations, delayed starts, and an evolution driven more by concerns for safety than improvements in efficacy.

BASIC PHARMACOLOGY OF LOCAL ANESTHETICS Chemistry Most local anesthetic agents consist of a lipophilic group (eg, an aromatic ring) connected by an intermediate chain via an ester or amide to an ionizable group (eg, a tertiary amine) (Table 26–1). In addition to the general physical properties of the molecules, specific stereochemical configurations are associated with differences in the potency of stereoisomers (eg, levobupivacaine, ropivacaine). Because ester links are more prone to hydrolysis than amide links, esters usually have a shorter duration of action. TABLE 26–1 Structure and properties of some ester and amide local anesthetics.1



Local anesthetics are weak bases and are usually made available clinically as salts to increase solubility and stability. In the body, they exist either as the uncharged base or as a cation (see Chapter 1, Ionization of Weak Acids and Weak Bases). The relative proportions of these two forms are governed by their pKa and the pH of the body fluids according to the Henderson-Hasselbalch equation, which can be expressed as: pKa = pH – log [base]/[conjugate acid] If the concentration of base and conjugate acid are equal, the second portion of the right side of the equation drops out, as log 1 = 0, leaving: pKa = pH (when base concentration = conjugate acid concentration)

Historical Development of Local Anesthesia Although the numbing properties of cocaine were recognized for centuries, one might consider September 15, 1884, to mark the “birth of local anesthesia.” Based on work performed by Carl Koller, cocaine’s numbing effect on the cornea was demonstrated before the Ophthalmological Congress in Heidelberg, ushering in the era of surgical local anesthesia. Unfortunately, with widespread use came recognition of cocaine’s significant CNS and cardiac toxicity, which along with its addiction potential, tempered enthusiasm for this application. As the early investigator Mattison commented, “the risk of untoward results have robbed this peerless drug of much favor in the minds of many surgeons, and so deprived them of a most valued ally.” As cocaine was known to be a benzoic acid ester, the search for alternative local anesthetics focused on this class of compounds, resulting in the identification of benzocaine shortly before the turn of the last century. However, benzocaine proved to have limited utility due to its marked hydrophobicity, and was thus relegated to topical anesthesia, a use for which it still finds limited application in current clinical practice. The first useful injectable local anesthetic, procaine, was introduced shortly thereafter by Einhorn, and its structure has served as the template for the development of the most commonly used modern local anesthetics. The three basic structural elements of these compounds can be appreciated by review of Table 26–1: an aromatic ring, conferring lipophilicity, an ionizable tertiary amine, conferring hydrophilicity, and an intermediate chain connecting these via an ester or amide linkage. One of procaine’s limitations was its short duration of action, a drawback overcome with the introduction of tetracaine in 1928. Unfortunately, tetracaine demonstrated significant toxicity when employed for high-volume peripheral blocks, ultimately reducing its common usage to spinal anesthesia. Both procaine and tetracaine shared another drawback: their ester linkage conferred instability, and particularly in the case of procaine, the free aromatic acid released during ester hydrolysis of the parent compound was believed to be the source of relatively frequent allergic reactions. Löfgren and Lundqvist circumvented the problem of instability with the introduction of lidocaine in 1948. Lidocaine was the first in a series of amino-amide local anesthetics that would come to dominate the second half of the 20th century. Lidocaine had a more favorable duration of action than procaine, and less systemic toxicity than tetracaine. To this day, it remains one of the most versatile and widely used anesthetics. Nonetheless, some applications required more prolonged block than that afforded by lidocaine, a pharmacologic void that was filled with the introduction of bupivacaine, a more lipophilic and more potent anesthetic. Unfortunately, bupivacaine was found to have greater propensity for significant effects on cardiac conduction and function, which at times proved lethal. Recognition of this potential for cardiac toxicity led to changes in anesthetic practice, and significant toxicity became sufficiently rare for it to remain a widely used anesthetic for nearly every regional technique in modern clinical practice. Nonetheless, this inherent cardiotoxicity would drive developmental work leading to the introduction of two recent additions to the anesthetic armamentarium, levobupivacaine and ropivacaine. The former is the S(–) enantiomer of bupivacaine, which has less affinity for cardiac sodium channels than its R(+) counterpart. Ropivacaine, another S(–) enantiomer, shares this reduced affinity for cardiac sodium channels, while being slightly less potent than bupivacaine or levobupivacaine. Thus, pKa can be seen as an effective way to consider the tendency for compounds to exist in a charged or uncharged form, ie, the lower the pKa, the greater the percentage of uncharged weak bases at a given pH. Because the pKa of most local anesthetics is in the range of 7.5–9.0, the charged, cationic form will constitute the larger percentage at physiologic pH. A glaring exception is benzocaine, which has a pKa around 3.5, and thus exists solely as the nonionized base under normal physiologic conditions. This issue of ionization is of critical importance because the cationic form is the most active at the receptor site. However, the story is a bit more complex, because the receptor site for local anesthetics is at the inner vestibule of the sodium channel, and the charged form of the anesthetic penetrates biologic membranes poorly. Thus, the uncharged form is important for cell penetration. After penetration into the cytoplasm, equilibration leads to formation and binding of the charged cation at the sodium channel, and hence the production of a clinical effect (Figure 26–1). Drug may also reach the receptor laterally through what has been termed the hydrophobic pathway. As a clinical consequence, local anesthetics are less effective when they are injected into infected tissues because the low extracellular pH favors the charged form, with less of the neutral base available for diffusion across the membrane. Conversely, adding bicarbonate to a


local anesthetic—a strategy sometimes utilized in clinical practice—will raise the effective concentration of the nonionized form and thus shorten the onset time of a regional block.

FIGURE 26–1 Schematic diagram depicting paths of local anesthetic (LA) to receptor sites. Extracellular anesthetic exists in equilibrium between charged and uncharged forms. The charged cation penetrates lipid membranes poorly; intracellular access is thus achieved by passage of the uncharged form. Intracellular re-equilibration results in formation of the more active charged species, which binds to the receptor at the inner vestibule of the sodium channel. Anesthetic may also gain access more directly by diffusing laterally within the membrane (hydrophobic pathway).

Pharmacokinetics When local anesthetics are used for local, peripheral, and central neuraxial anesthesia—their most common clinical applications— systemic absorption, distribution, and elimination serve only to diminish or terminate their effect. Thus, classic pharmacokinetics plays a lesser role than with systemic therapeutics, yet remains important to the anesthetic’s duration and critical to the potential development of adverse reactions, specifically cardiac and central nervous system (CNS) toxicity. Some pharmacokinetic properties of the commonly used amide local anesthetics are summarized in Table 26–2. The pharmacokinetics of the ester-based local anesthetics has not been extensively studied owing to their rapid breakdown in plasma (elimination half-life < 1 minute). TABLE 26–2 Pharmacokinetic properties of several amide local anesthetics.

A. Absorption Systemic absorption of injected local anesthetic from the site of administration is determined by several factors, including dosage, site of injection, drug-tissue binding, local tissue blood flow, use of a vasoconstrictor (eg, epinephrine), and the physicochemical properties of the


drug itself. Anesthetics that are more lipid soluble are generally more potent, have a longer duration of action, and take longer to achieve their clinical effect. Extensive protein binding also serves to increase the duration of action. Application of a local anesthetic to a highly vascular area such as the tracheal mucosa or the tissue surrounding intercostal nerves results in more rapid absorption and thus higher blood levels than if the local anesthetic is injected into a poorly perfused tissue such as subcutaneous fat. When used for major conduction blocks, the peak serum levels will vary as a function of the specific site of injection, with intercostal blocks among the highest, and sciatic and femoral among the lowest (Figure 26–2). When vasoconstrictors are used with local anesthetics, the resultant reduction in blood flow serves to reduce the rate of systemic absorption and thus diminishes peak serum levels. This effect is generally most evident with the shorter-acting, less potent, and less lipid-soluble anesthetics.

FIGURE 26–2 Comparative peak blood levels of several local anesthetic agents following administration into various anatomic sites. (Adapted, with permission, from Covino BD, Vassals HG: Local Anesthetics: Mechanism of Action in Clinical Use. Grune & Stratton, 1976. Copyright Elsevier.)

B. Distribution 1. Localized—As local anesthetic is usually injected directly at the site of the target organ, distribution within this compartment plays an essential role with respect to achievement of clinical effect. For example, anesthetics delivered into the subarachnoid space will be diluted with cerebrospinal fluid (CSF) and the pattern of distribution will be dependent upon a host of factors, among the most critical


being the specific gravity relative to that of CSF and the patient’s position. Solutions are termed hyperbaric, isobaric, and hypobaric, and will respectively descend, remain relatively static, or ascend, within the subarachnoid space due to gravity when the patient sits upright. A review and analysis of relevant literature cited 25 factors that have been invoked as determinants of spread of local anesthetic in CSF, which can be broadly classified as characteristics of the anesthetic solution, CSF constituents, patient characteristics, and techniques of injection. Somewhat similar considerations apply to epidural and peripheral blocks. 2. Systemic—The peak blood levels achieved during major conduction anesthesia will be minimally affected by the concentration of anesthetic or the speed of injection. The disposition of these agents can be well approximated by a two-compartment model. The initial alpha phase reflects rapid distribution in blood and highly perfused organs (eg, brain, liver, heart, kidney), characterized by a steep exponential decline in concentration. This is followed by a slower declining beta phase reflecting distribution into less well perfused tissue (eg, muscle, gut), and may assume a nearly linear rate of decline. The potential toxicity of the local anesthetics is affected by the protective effect afforded by uptake by the lungs, which serve to attenuate the arterial concentration, though the time course and magnitude of this effect have not been adequately characterized. C. Metabolism and Excretion The local anesthetics are converted to more water-soluble metabolites in the liver (amide type) or in plasma (ester type), which are excreted in the urine. Since local anesthetics in the uncharged form diffuse readily through lipid membranes, little or no urinary excretion of the neutral form occurs. Acidification of urine promotes ionization of the tertiary amine base to the more water-soluble charged form, leading to more rapid elimination. Ester-type local anesthetics are hydrolyzed very rapidly in the blood by circulating butyrylcholinesterase to inactive metabolites. For example, the half-lives of procaine and chloroprocaine in plasma are less than a minute. However, excessive concentrations may accumulate in patients with reduced or absent plasma hydrolysis secondary to atypical plasma cholinesterase. The amide local anesthetics undergo complex biotransformation in the liver, which includes hydroxylation and N-dealkylation by liver microsomal cytochrome P450 isozymes. There is considerable variation in the rate of liver metabolism of individual amide compounds, with prilocaine (fastest) > lidocaine > mepivacaine > ropivacaine ≈ bupivacaine and levobupivacaine (slowest). As a result, toxicity from amide-type local anesthetics is more likely to occur in patients with hepatic disease. For example, the average elimination half-life of lidocaine may be increased from 1.6 hours in normal patients (t½, Table 26–2) to more than 6 hours in patients with severe liver disease. Many other drugs used in anesthesia are metabolized by the same P450 isozymes, and concomitant administration of these competing drugs may slow the hepatic metabolism of the local anesthetics. Decreased hepatic elimination of local anesthetics would also be anticipated in patients with reduced hepatic blood flow. For example, the hepatic elimination of lidocaine in patients anesthetized with volatile anesthetics (which reduce liver blood flow) is slower than in patients anesthetized with intravenous anesthetic techniques. Delayed metabolism due to impaired hepatic blood flow may likewise occur in patients with congestive heart failure.

Pharmacodynamics A. Mechanism of Action 1. Membrane potential—The primary mechanism of action of local anesthetics is blockade of voltage-gated sodium channels (Figure 26–1). The excitable membrane of nerve axons, like the membrane of cardiac muscle (see Chapter 14) and neuronal cell bodies (see Chapter 21), maintains a resting transmembrane potential of –90 to –60 mV. During excitation, the sodium channels open, and a fast, inward sodium current quickly depolarizes the membrane toward the sodium equilibrium potential (+40 mV). As a result of this depolarization process, the sodium channels close (inactivate) and potassium channels open. The outward flow of potassium repolarizes the membrane toward the potassium equilibrium potential (about –95 mV); repolarization returns the sodium channels to the rested state with a characteristic recovery time that determines the refractory period. The transmembrane ionic gradients are maintained by the sodium pump. These ionic fluxes are similar to, but simpler than, those in heart muscle, and local anesthetics have similar effects in both tissues. 2. Sodium channel isoforms—Each sodium channel consists of a single alpha subunit containing a central ion-conducting pore associated with accessory beta subunits. The pore-forming alpha subunit is actually sufficient for functional expression, but the kinetics and voltage dependence of channel gating are modified by the beta subunit. A variety of different sodium channels have been characterized by electrophysiologic recording, and subsequently isolated and cloned, while mutational analysis has allowed for identification of the essential components of the local anesthetic binding site. Nine members of a mammalian family of sodium channels have been so characterized and classified as Nav 1.1–Nav 1.9, where the chemical symbol represents the primary ion, the subscript denotes the physiologic regulator (in this case voltage), the initial number denotes the gene, and the number following the period indicates the particular isoform. 3. Channel blockade—Biologic toxins such as batrachotoxin, aconitine, veratridine, and some scorpion venoms bind to receptors within the channel and prevent inactivation. This results in prolonged influx of sodium through the channel and depolarization of the resting potential. The marine toxins tetrodotoxin (TTX) and saxitoxin have clinical effects that largely resemble those of local anesthetics (ie,


block of conduction without a change in the resting potential). However, in contrast to the local anesthetics, their binding site is located near the extracellular surface. The sensitivity of these channels to TTX varies, and subclassification based on this pharmacologic sensitivity has important physiologic and therapeutic implications. Six of the aforementioned channels are sensitive to nanomolar concentration of this biotoxin (TTX-S), while three are resistant (TTX-R). Of the latter, Na v 1.8 and Nav 1.9 appear to be exclusively expressed in dorsal root ganglia nociceptors, which raises the developmental possibility of targeting these specific neuronal subpopulations. Such fine-tuned analgesic therapy has the theoretical potential of providing effective analgesia, while limiting the significant adverse effects produced by nonspecific sodium channel blockers. When progressively increasing concentrations of a local anesthetic are applied to a nerve fiber, the threshold for excitation increases, impulse conduction slows, the rate of rise of the action potential declines, action potential amplitude decreases, and, finally, the ability to generate an action potential is completely abolished. These progressive effects result from binding of the local anesthetic to more and more sodium channels. If the sodium current is blocked over a critical length of the nerve, propagation across the blocked area is no longer possible. In myelinated nerves, the critical length appears to be two to three nodes of Ranvier. At the minimum dose required to block propagation, the resting potential is not significantly altered. The blockade of sodium channels by most local anesthetics is both voltage and time dependent: Channels in the rested state, which predominate at more negative membrane potentials, have a much lower affinity for local anesthetics than activated (open state) and inactivated channels, which predominate at more positive membrane potentials (see Figure 14–10). Therefore, the effect of a given drug concentration is more marked in rapidly firing axons than in resting fibers (Figure 26–3). Between successive action potentials, a portion of the sodium channels will recover from the local anesthetic block (see Figure 14–10). The recovery from drug-induced block is 10– 1000 times slower than the recovery of channels from normal inactivation (as shown for the cardiac membrane in Figure 14–4). As a result, the refractory period is lengthened and the nerve conducts fewer action potentials.

FIGURE 26–3 Effect of repetitive activity on the block of sodium current produced by a local anesthetic in a myelinated axon. A series of 25 pulses was applied, and the resulting sodium currents (downward deflections) are superimposed. Note that the current produced by the pulses rapidly decreased from the first to the 25th pulse. A long rest period after the train resulted in recovery from block, but the block could be reinstated by a subsequent train. nA, nanoamperes. (Adapted, with permission, from Courtney KR: Mechanism of frequency-dependent inhibition of sodium currents in frog myelinated nerve by the lidocaine derivative GEA. J Pharmacol Exp T her 1975;195:225.)

Elevated extracellular calcium partially antagonizes the action of local anesthetics owing to the calcium-induced increase in the surface potential on the membrane (which favors the low-affinity rested state). Conversely, increases in extracellular potassium depolarize the membrane potential and favor the inactivated state, enhancing the effect of local anesthetics. 4. Other effects—Currently used local anesthetics bind to the sodium channel with low affinity and poor specificity, and there are multiple other sites for which their affinity is nearly the same as that for sodium channel binding. Thus, at clinically relevant concentrations, local anesthetics are potentially active at countless other channels (eg, potassium and calcium), enzymes (eg, adenylyl cyclase, carnitine-acylcarnitine translocase), and receptors (eg, N-methyl-D-aspartate [NMDA], G protein-coupled, 5-HT3 , neurokinin-1 [substance P receptor]). The role that such ancillary effects play in achievement of local anesthesia appears to be important but is poorly understood. Further, interactions with these other sites are likely the basis for numerous differences between the local anesthetics with respect to anesthetic effects (eg, differential block) and toxicities that do not parallel anesthetic potency, and thus are not adequately accounted for solely by blockade of the voltage-gated sodium channel. The actions of circulating local anesthetics at such diverse sites exert a multitude of effects, some of which go beyond pain control, including some that are also potentially beneficial. For example, there is evidence to suggest that the blunting of the stress response and


improvements in perioperative outcome that may occur with epidural anesthesia derive in part from an action of the anesthetic beyond its sodium channel block. Circulating anesthetics also demonstrate antithrombotic effects having an impact on coagulation, platelet aggregation, and the microcirculation, as well as modulation of inflammation. B. Structure-Activity Characteristics of Local Anesthetics The smaller and more highly lipophilic local anesthetics have a faster rate of interaction with the sodium channel receptor. As previously noted, potency is also positively correlated with lipid solubility. Lidocaine, procaine, and mepivacaine are more water soluble than tetracaine, bupivacaine, and ropivacaine. The latter agents are more potent and have longer durations of local anesthetic action. These long-acting local anesthetics also bind more extensively to proteins and can be displaced from these binding sites by other protein-bound drugs. In the case of optically active agents (eg, bupivacaine), the R(+) isomer can usually be shown to be slightly more potent than the S(–) isomer (levobupivacaine). C. Neuronal Factors Affecting Block 1. Differential block—Since local anesthetics are capable of blocking all nerves, their actions are not limited to the desired loss of sensation from sites of noxious (painful) stimuli. With central neuraxial techniques (spinal or epidural), motor paralysis may impair respiratory activity, and autonomic nerve blockade may promote hypotension. Further, while motor paralysis may be desirable during surgery, it may be a disadvantage in other settings. For example, motor weakness occurring as a consequence of epidural anesthesia during obstetrical labor may limit the ability of the patient to bear down (ie, “push”) during delivery. Similarly, when used for postoperative analgesia, weakness may hamper ability to ambulate without assistance and pose a risk of falling, while residual autonomic blockade may interfere with bladder function, resulting in urinary retention and the need for bladder catheterization. These issues are particularly problematic in the setting of ambulatory (same-day) surgery, which represents an ever-increasing percentage of surgical caseloads. 2. Intrinsic susceptibility of nerve fibers—Nerve fibers differ significantly in their susceptibility to local anesthetic blockade. It has been traditionally taught, and still often cited, that local anesthetics preferentially block smaller diameter fibers first because the distance over which such fibers can passively propagate an electrical impulse is shorter. However, a variable proportion of large fibers are blocked prior to the disappearance of the small fiber component of the compound action potential. Most notably, myelinated nerves tend to be blocked before unmyelinated nerves of the same diameter. For example, preganglionic B fibers are blocked before the smaller unmyelinated C fibers involved in pain transmission (Table 26–3). TABLE 26–3 Relative size and susceptibility of different types of nerve fibers to local anesthetics.


Another important factor underlying differential block derives from the state- and use-dependent mechanism of action of local anesthetics. Blockade by these drugs is more marked at higher frequencies of depolarization. Sensory (pain) fibers have a high firing rate and relatively long action potential duration. Motor fibers fire at a slower rate and have a shorter action potential duration. As type A delta and C fibers participate in high-frequency pain transmission, this characteristic may favor blockade of these fibers earlier and with lower concentrations of local anesthetics. The potential impact of such effects mandates cautious interpretation of non-physiologic experiments evaluating intrinsic susceptibility of nerves to conduction block by local anesthetics. 3. Anatomic arrangement—In addition to the effect of intrinsic vulnerability to local anesthetic block, the anatomic organization of the peripheral nerve bundle may impact the onset and susceptibility of its components. As one would predict based on the necessity of having proximal sensory fibers join the nerve trunk last, the core will contain sensory fibers innervating the most distal sites. Anesthetic placed outside the nerve bundle will thus reach and anesthetize the proximal fibers located at the outer portion of the bundle first, and sensory block will occur in sequence from proximal to distal.

CLINICAL PHARMACOLOGY OF LOCAL ANESTHETICS Local anesthetics can provide highly effective analgesia in well-defined regions of the body. The usual routes of administration include topical application (eg, nasal mucosa, wound [incision site] margins), injection in the vicinity of peripheral nerve endings (perineural infiltration) and major nerve trunks (blocks), and injection into the epidural or subarachnoid spaces surrounding the spinal cord (Figure 26–4).



FIGURE 26–4 Schematic diagram of the typical sites of injection of local anesthetics in and around the spinal canal. When local anesthetics are injected extradurally, it is referred to as an epidural block. A caudal block is a specific type of epidural block in which a needle is inserted into the caudal canal via the sacral hiatus. Injections around peripheral nerves are known as perineural blocks (eg, paravertebral block). Finally, injection into cerebrospinal fluid in the subarachnoid (intrathecal) space is referred to as a spinal block.

Clinical Block Characteristics In clinical practice, there is generally an orderly evolution of block components beginning with sympathetic transmission and progressing to temperature, pain, light touch, and finally motor block. This is most readily appreciated during onset of spinal anesthesia, where a spatial discrepancy can be detected in modalities, the most vulnerable components achieving greater dermatomal (cephalad) spread. Thus, loss of the sensation of cold (often assessed by a wet alcohol sponge) will be roughly two segments above the analgesic level for pinprick, which in turn will be roughly two segments rostral to loss of light touch recognition. However, because of the anatomic considerations noted earlier for peripheral nerve trunks, onset with peripheral blocks is more variable, and proximal motor weakness may precede onset of more distal sensory loss. Additionally, anesthetic solution is not generally deposited evenly around a nerve bundle, and longitudinal spread and radial penetration into the nerve trunk are far from uniform. With respect to differential block, it is worth noting that “successful” surgical anesthesia may require loss of touch, not just ablation of pain, as some patients will find even the sensation of touch distressing during surgery, often fearing that the procedure may become painful. Further, while differences may exist in modalities, it is not possible with conventional techniques to produce surgical anesthesia without some loss of motor function. A. Effect of Added Vasoconstrictors Several benefits may be derived from addition of a vasoconstrictor to a local anesthetic. First, localized neuronal uptake is enhanced because of higher sustained local tissue concentrations that can translate clinically into a longer duration block. This may enable adequate anesthesia for more prolonged procedures, extended duration of postoperative pain control, and lower total anesthetic requirement. Second, peak blood levels will be lowered as absorption is more closely matched to metabolism and elimination, and the risk of systemic toxic effects is reduced. Moreover, when incorporated into a spinal anesthetic, epinephrine may not only contribute to prolongation of the local anesthetic effect via its vasoconstrictor properties, but also exert a direct analgesic effect mediated by postsynaptic α2 adrenoceptors within the spinal cord. Recognition of this potential has led to the clinical use of the α2 agonist clonidine as a local anesthetic adjuvant for spinal anesthesia. Conversely, inclusion of epinephrine may have untoward effects. The addition of epinephrine to anesthetic solutions can potentiate the neurotoxicity of local anesthetics used for peripheral nerve blocks or spinal anesthesia. Further, the use of a vasoconstrictor agent in an area that lacks adequate collateral flow (eg, digital block) is generally avoided, though some have questioned the validity of this proscription. B. Intentional Use of Systemic Local Anesthetics Although the principal use of local anesthetics is to achieve anesthesia in a restricted area, these agents are sometimes deliberately administered systemically to take advantage of suppressive effects on pain processing. In addition to documented reductions in anesthetic requirement and postoperative pain, systemic administration of local anesthetics has been used with some success in the treatment of chronic pain, and this effect may outlast the duration of anesthetic exposure. The achievement of pain control by systemic administration of local anesthetics is thought to derive, at least in part, from the suppression of abnormal ectopic discharge, an effect observed at concentrations of local anesthetic an order of magnitude lower than those required for blockade of propagation of action potentials in normal nerves. Consequently, these effects can be achieved without the adverse effects that would derive from failure of normal nerve conduction. Escalating doses of anesthetic appear to exert the following systemic actions: (1) low concentrations may preferentially suppress ectopic impulse generation in chronically injured peripheral nerves; (2) moderate concentrations may suppress central sensitization, which would explain therapeutic benefit that may extend beyond the anesthetic exposure; and (3) higher concentrations will produce general analgesic effects and may culminate in serious toxicity.

Toxicity Local anesthetic toxicity derives from two distinct processes: (1) systemic effects following inadvertent intravascular injection or absorption of the local anesthetic from the site of administration; and (2) neurotoxicity resulting from local effects produced by direct contact with neural elements. A. Systemic Toxicity The dose of local anesthetic used for epidural anesthesia or high-volume peripheral blocks is sufficient to produce major clinical toxicity, even death. To minimize risk, maximum recommended doses for each drug for each general application have been promulgated. The


concept underlying this approach is that absorption from the site of injection should appropriately match metabolism, thereby preventing toxic serum levels. However, these recommendations do not consider patient characteristics or concomitant risk factors, nor do they take into account the specific peripheral nerve block performed, which has a significant impact on the rate of systemic uptake (Figure 26–2). Most importantly, they fail to afford protection from toxicity induced by inadvertent intravascular injection (occasionally into an artery, but more commonly a vein). 1. CNS toxicity—All local anesthetics have the ability to produce sedation, light-headedness, visual and auditory disturbances, and restlessness when high plasma concentrations result from rapid absorption or inadvertent intravascular administration. An early symptom of local anesthetic toxicity is circumoral and tongue numbness and a metallic taste. At higher concentrations, nystagmus and muscular twitching occur, followed by tonic-clonic convulsions. Local anesthetics apparently cause depression of cortical inhibitory pathways, thereby allowing unopposed activity of excitatory neuronal pathways. This transitional stage of unbalanced excitation (ie, seizure activity) is then followed by generalized CNS depression. However, this classic pattern of evolving toxicity has been largely characterized in human volunteer studies (which are ethically constrained to low doses), and by graded administration in animal models. Deviations from such classic progression are common in clinical toxicity and will be influenced by a host of factors, including patient vulnerability, the particular anesthetic administered, concurrent drugs, and rate of rise of serum drug levels. A recent literature review of reported clinical cases of local anesthetic cardiac toxicity found prodromal signs of CNS toxicity in only 18% of cases. When large doses of a local anesthetic are required (eg, for major peripheral nerve block or local infiltration for major plastic surgery), premedication with a parenteral benzodiazepine (eg, diazepam or midazolam) will provide some prophylaxis against local anesthetic-induced CNS toxicity. However, such premedication will have little, if any, effect on cardiovascular toxicity, potentially delaying recognition of a life-threatening overdose. Of note, administration of a propofol infusion or general anesthesia accounted for 5 of the 10 cases presenting with isolated cardiovascular toxicity in the aforementioned literature review of reported clinical cases. If seizures do occur, it is critical to prevent hypoxemia and acidosis, which potentiate anesthetic toxicity. Rapid tracheal intubation can facilitate adequate ventilation and oxygenation, and is essential to prevent pulmonary aspiration of gastric contents in patients at risk. The effect of hyperventilation is complex, and its role in resuscitation following anesthetic overdose is somewhat controversial, but it likely offers distinct benefit if used to counteract metabolic acidosis. Seizures induced by local anesthetics should be rapidly controlled to prevent patient harm and exacerbation of acidosis. A recent practice advisory from the American Society of Regional Anesthesia advocates benzodiazepines as first-line drugs (eg, midazolam, 0.03–0.06 mg/kg) because of their hemodynamic stability, but small doses of propofol (eg, 0.25–0.5 mg/kg) were considered acceptable alternatives, as they are often more immediately available in the setting of local anesthetic administration. The motor activity of the seizure can be effectively terminated by administration of a neuromuscular blocker, though this will not diminish the CNS manifestations, and efforts must include therapy directed at the underlying seizure activity. 2. Cardiotoxicity—The most feared complications associated with local anesthetic administration result from the profound effects these agents can have on cardiac conduction and function. In 1979, an editorial by Albright reviewed the circumstances of six deaths associated with the use of bupivacaine and etidocaine. This seminal publication suggested that these relatively new lipophilic and potent anesthetics had greater potential cardiotoxicity, and that cardiac arrest could occur concurrently or immediately following seizures and, most importantly, in the absence of hypoxia or acidosis. Although this suggestion was sharply criticized, subsequent clinical experience unfortunately reinforced Albright’s concern—within 4 years the FDA had received reports of 12 cases of cardiac arrest associated with the use of 0.75% bupivacaine for epidural anesthesia in obstetrics. Further support for enhanced cardiotoxicity of these anesthetics came from animal studies demonstrating that doses of bupivacaine and etidocaine as low as two thirds those producing convulsions could induce arrhythmias, while the margin between CNS and cardiac toxicity was less than half that for lidocaine. In response, the FDA banned the use of 0.75% bupivacaine in obstetrics. In addition, incorporation of a test dose became ingrained as a standard of anesthetic practice, along with the practice of fractionated administration of local anesthetic. Although reduction in bupivacaine’s anesthetic concentration and changes in anesthetic practice did much to reduce the risk of cardiotoxicity, the recognized differences in the toxicity of the stereoisomers comprising bupivacaine created an opportunity for the development of potentially safer anesthetics (see Chapter 1). Investigations demonstrated that the enantiomers of the racemic mixture bupivacaine were not equivalent with respect to cardiotoxicity, the S(–) enantiomer having better therapeutic advantage, leading to the subsequent marketing of levobupivacaine. This was followed shortly thereafter by ropivacaine, a slightly less potent anesthetic than bupivacaine. It should be noted, however, that the reduction in toxicity afforded by these compounds is only modest, and that risk of significant cardiotoxicity remains a very real concern when these anesthetics are administered for high-volume blocks. 3. Reversal of bupivacaine toxicity—Recently, a series of clinical events, serendipitous observations, systematic experimentation, and astute clinical decisions have identified a relatively simple, practical and apparently effective therapy for resistant bupivacaine cardiotoxicity using intravenous infusion of lipid. Furthermore, this therapy appears to have applications that extend beyond bupivacaine cardiotoxicity to the cardiac or CNS toxicity induced by an overdose of any lipid-soluble drug (see Box: Lipid Resuscitation). B. Localized Toxicity 1. Neural injury—From the early introduction of spinal anesthesia into clinical practice, sporadic reports of neurologic injury associated


with this technique raised concern that local anesthetic agents were potentially neurotoxic. Following injuries associated with Durocaine —a spinal anesthetic formulation containing procaine—initial attention focused on the vehicle components. However, experimental studies found 10% procaine alone induced similar injuries in cats, whereas the vehicle did not. Concern for anesthetic neurotoxicity reemerged in the early 1980s with a series of reports of major neurologic injury occurring with the use of chloroprocaine for epidural anesthesia. In these cases, there was evidence that anesthetic intended for the epidural space was inadvertently administered intrathecally. As the dose required for spinal anesthesia is roughly an order of magnitude less than for epidural anesthesia, injury was apparently the result of excessive exposure of the more vulnerable subarachnoid neural elements. With changes in vehicle formulation and in clinical practice, concern for toxicity again subsided, only to reemerge a decade later with reports of cauda equina syndrome associated with continuous spinal anesthesia (CSA). In contrast to the more common single-injection technique, CSA involves placing a catheter in the subarachnoid space to permit repetitive dosing to facilitate adequate anesthesia and maintenance of block for extended periods. In these cases the local anesthetic was evidently administered to a relatively restricted area of the subarachnoid space; in order to extend the block to achieve adequate surgical anesthesia, multiple repetitive doses of anesthetic were then administered. By the time the block was adequate, neurotoxic concentrations had accumulated in a restricted area of the caudal region of the subarachnoid space. Most notably, the anesthetic involved in the majority of these cases was lidocaine, a drug most clinicians considered to be the least toxic of agents. This was followed by reports of neurotoxic injury occurring with lidocaine intended for epidural administration that had inadvertently been administered intrathecally, similar to the cases involving chloroprocaine a decade earlier. The occurrence of neurotoxic injury with CSA and subarachnoid administration of epidural doses of lidocaine served to establish vulnerability whenever excessive anesthetic was administered intrathecally, regardless of the specific anesthetic used. Of even more concern, subsequent reports provided evidence for injury with spinal lidocaine administered at the high end of the recommended clinical dosage, prompting recommendations for a reduction in maximum dose. These clinical reports (as well as concurrent experimental studies) served to dispel the concept that modern local anesthetics administered at clinically relevant doses and concentrations were incapable of inducing neurotoxic injury. The mechanism of local anesthetic neurotoxicity has been extensively investigated in cell culture, isolated axons, and in vivo models. These studies have demonstrated myriad deleterious effects including conduction failure, membrane damage, enzyme leakage, cytoskeletal disruption, accumulation of intracellular calcium, disruption of axonal transport, growth cone collapse, and apoptosis. It is not clear what role these factors or others play in clinical injury. It is clear, however, that injury does not result from blockade of the voltagegated sodium channel per se, and thus clinical effect and toxicity are not tightly linked.

Lipid Resuscitation Based on a case of apparent cardiotoxicity from a very low dose of bupivacaine in a patient with carnitine deficiency, Weinberg postulated that this metabolic derangement led to enhanced toxicity due to the accumulation of fatty acids within the cardiac myocyte. He hypothesized that administration of lipid would similarly potentiate bupivacaine cardiotoxicity, but experiments performed to test this hypothesis demonstrated exactly the opposite effect. Accordingly, he began systematic laboratory investigations, which clearly demonstrated the potential efficacy of an intravenous lipid emulsion (ILE) for resuscitation from bupivacaine cardiotoxicity. Clinical confirmation came 8 years later with the report of the successful resuscitation of a patient who sustained an anesthetic-induced (bupivacaine plus mepivacaine) cardiac arrest refractory to standard advanced cardiac life support procedures (ACLS). Numerous similar reports of successful resuscitations soon followed, extending this clinical experience to other anesthetics including levobupivacaine and ropivacaine, anesthetic-induced CNS toxicity, as well as toxicity induced by other classes of compounds, eg, bupropion-induced cardiovascular collapse and multiform ventricular tachycardia provoked by haloperidol. Laboratory investigations have likewise provided evidence of efficacy for treatment of diverse toxic challenges (eg, verapamil, clomipramine, and propranolol). The mechanism by which lipid is effective is incompletely understood, but almost certainly some of its effect is related to its ability to extract a lipophilic drug from aqueous plasma and thus reducing its effective concentration at tissue targets, a mechanism termed “lipid sink.” However, the extent of this extraction does not appear adequate to account for the magnitude of clinical effect, suggesting that other mechanisms at least contribute to the efficacy of lipid rescue. For example, bupivacaine has been shown to inhibit fatty acid transport at the inner mitochondrial membrane, and lipid might act by overcoming this inhibition serving to restore energy to the myocardium or derive benefit via elevation of intramyocyte calcium concentration. Although numerous questions remain, the evolving evidence is sufficient to warrant administration of lipid in cases of systemic anesthetic toxicity. Its use has been promulgated by a task force of the American Society of Regional Anesthesia, and administration of lipid has been incorporated into the most recent revision of ACLS guidelines for Cardiac Arrest in Special Situations. Importantly, propofol cannot be administered for this purpose, as the relatively enormous volume of this solution required for lipid therapy would deliver lethal quantities of propofol. 2. Transient neurologic symptoms (TNS)—In addition to the very rare but devastating neural complications that can occur with neuraxial (spinal and epidural) administration of local anesthetics, a syndrome of transient pain or dysesthesia, or both, has been recently


linked to use of lidocaine for spinal anesthesia. Although these symptoms are not associated with sensory loss, motor weakness, or bowel and bladder dysfunction, the pain can be quite severe, often exceeding that induced by the surgical procedure. TNS occurs even at modest doses of anesthetic, and has been documented in as many as one third of patients receiving lidocaine, with increased risk associated with certain patient positions during surgery (eg, lithotomy), and with ambulatory anesthesia. Risk with other anesthetics varies considerably. For example, the incidence is only slightly reduced with procaine or mepivacaine but appears to be negligible with bupivacaine, prilocaine, and chloroprocaine. The etiology and significance of TNS remain to be established, but differences between factors affecting TNS and experimental animal toxicity argue strongly against a common mechanism mediating these symptoms and persistent or permanent neurologic deficits. Nonetheless, the high incidence of TNS has greatly contributed to dissatisfaction with lidocaine as a spinal anesthetic, leading to its near abandonment for this technique (although it remains a popular and appropriate anesthetic for all other applications, including epidural anesthesia). Chloroprocaine, once considered a more toxic anesthetic, is now being explored for short-duration spinal anesthesia as an alternative to lidocaine, a compound that has been used for well over 50 million spinal anesthetic procedures.

COMMONLY USED LOCAL ANESTHETICS & THEIR APPLICATIONS ARTICAINE Approved for use in the USA as a dental anesthetic in April 2000, articaine is unique among the amino-amide anesthetics in having a thiophene, rather than a benzene ring, as well as an additional ester group that is subject to metabolism by plasma esterases (Table 26–1). The modification of the ring serves to enhance lipophilicity, and thus improve tissue penetration, while inclusion of the ester leads to a shorter plasma half-life (approximately 20 minutes) potentially imparting a better therapeutic index with respect to systemic toxicity. These characteristics have led to widespread popularity in dental anesthesia, where it is generally considered to be more effective, and possibly safer, than lidocaine, the prior standard. Balanced against these positive attributes are concerns that development of persistent paresthesias, while rare, may be three times more common with articaine. However, prilocaine has been associated with an even higher relative incidence (twice that of articaine). Importantly, these are the only two dental anesthetics that are formulated as 4% solutions; the others are all marketed at lower concentrations (eg, the maximum concentration of lidocaine used for dental anesthesia is 2%), and it is well established that anesthetic neurotoxicity is, to some extent, concentration-dependent. Thus, it is quite possible that enhanced risk derives from the formulation rather than from an intrinsic property of the anesthetic. In a recent survey of US and Canadian Dental Schools, over half of respondents indicated that 4% articaine is no longer used for mandibular nerve block.

BENZOCAINE As previously noted, benzocaine’s pronounced lipophilicity has relegated its application to topical anesthesia. However, despite over a century of use for this purpose, its popularity has recently diminished owing to increasing concerns regarding its potential to induce methemoglobinemia. Elevated levels can be due to inborn errors, or can occur with exposure to an oxidizing agent, and such is the case with significant exposure to benzocaine (or nitrites, see Chapter 12). Because methemoglobin does not transport oxygen, elevated levels pose serious risk, with severity obviously paralleling blood levels.

BUPIVACAINE Based on concerns for cardiotoxicity, bupivacaine is often avoided for techniques that demand high volumes of concentrated anesthetic, such as epidural or peripheral nerve blocks performed for surgical anesthesia. In contrast, relatively low concentrations (≤ 0.25%) are frequently used to achieve prolonged peripheral anesthesia and analgesia for postoperative pain control, and the drug enjoys similar popularity where anesthetic infiltration is used to control pain from a surgical incision. It is often the agent of choice for epidural infusions used for postoperative pain control and for labor analgesia. Finally, it has a comparatively unblemished record as a spinal anesthetic, with a relatively favorable therapeutic index with respect to neurotoxicity, and little, if any, risk of TNS. However, spinal bupivacaine is not well suited for outpatient or ambulatory surgery, because its relatively long duration of action can delay recovery, resulting in a longer stay prior to discharge to home.

CHLOROPROCAINE The introduction of chloroprocaine into clinical practice in 1951 represented a reversion to the earlier amino-ester template. Chloroprocaine gained widespread use as an epidural agent in obstetrical anesthesia where its rapid hydrolysis served to minimize risk of systemic toxicity or fetal exposure. The unfortunate reports of neurologic injury associated with apparent intrathecal misplacement of large doses intended for the epidural space led to its near abandonment. However, the frequent occurrence of TNS with lidocaine


administered as a spinal anesthetic has created an anesthetic void that chloroprocaine appears well suited to fill. The onset and duration of action of spinal chloroprocaine are even shorter than those of lidocaine, while presenting little, if any, risk of TNS. Although never exonerated with respect to the early neurologic injuries associated with epidural anesthesia, it is now appreciated that high doses of any local anesthetic are capable of inducing neurotoxic injury. A formulation is now marketed in Europe specifically for spinal anesthesia, and there is considerable off-label use of a preservative-free solution in the USA. Nonetheless, documented use as a spinal anesthetic is relatively limited, and additional experience will be required to firmly establish safety. In addition to chloroprocaine’s emerging use for spinal anesthesia, it still finds some current use as an epidural anesthetic, particularly in circumstances where there is an indwelling catheter and the need for quick attainment of surgical anesthesia, such as caesarian section for a laboring parturient with a compromised fetus.

COCAINE Current clinical use of cocaine is largely restricted to topical anesthesia for ear, nose, and throat procedures, where its intense vasoconstriction can serve to reduce bleeding. Even here, use has diminished in favor of other anesthetics combined with vasoconstrictors because of concerns about systemic toxicity, as well as the inconvenience of dispensing and handling this controlled substance.

ETIDOCAINE Introduced along with bupivacaine, etidocaine has had limited application due to its poor block characteristics. It has a tendency to produce an inverse differential block (ie, compared with other anesthetics such as bupivacaine, it produces excessive motor relative to sensory block), which is rarely a favorable attribute.

LEVOBUPIVACAINE As previously discussed, this S(–) enantiomer of bupivacaine is somewhat less cardiotoxic than the racemic mixture. It is also less potent, and tends to have a longer duration of action, though the magnitude of these effects is too small to have any substantial clinical significance. Interestingly, recent work with lipid resuscitation suggests a potential advantage of levobupivacaine over ropivacaine, as the former is more effectively sequestered into a so-called lipid sink, implying greater ability to reverse toxic effects should they occur.

LIDOCAINE Aside from the issue of a high incidence of TNS with spinal administration, lidocaine has had an excellent record as an intermediate duration anesthetic, and remains the reference standard against which most anesthetics are compared.

MEPIVACAINE Although structurally similar to bupivacaine and ropivacaine (Table 26–1), mepivacaine displays clinical properties that are comparable to lidocaine. However, it differs from lidocaine with respect to vasoactivity, as it has a tendency toward vasoconstriction rather than vasodilation. This characteristic likely accounts for its slightly longer duration of action, which has made it a popular choice for major peripheral blocks. Lidocaine has retained its dominance over mepivacaine for epidural anesthesia, where the routine placement of a catheter negates the importance of a longer duration. More importantly, mepivacaine is slowly metabolized by the fetus, making it a poor choice for epidural anesthesia in the parturient. When used for spinal anesthesia, mepivacaine has a slightly lower incidence of TNS than lidocaine.

PRILOCAINE Prilocaine has the highest clearance of the amino-amide anesthetics, imparting reduced risk of systemic toxicity. Unfortunately, this is somewhat offset by its propensity to induce methemoglobinemia, which results from accumulation of one its metabolites, ortho-toluidine, an oxidizing agent. As a spinal anesthetic, prilocaine’s duration of action is slightly longer than that of lidocaine, and the limited data suggest it carries a low risk of TNS. It is gaining increasing use for spinal anesthesia in Europe, where it has been marketed specifically for this purpose. No approved formulation exists in the USA, and there is no formulation that would be appropriate to use for spinal anesthesia as an off-label indication.


ROPIVACAINE Ropivacaine is an S(–) enantiomer in a homologous series that includes bupivacaine and mepivacaine, distinguished by its chirality, and the propyl group off the piperidine ring (Table 26–1). Its perceived reduced cardiotoxicity has led to widespread use for high-volume peripheral blocks. It is also a popular choice for epidural infusions for control of labor and postoperative pain. Although there is some evidence to suggest that ropivacaine might produce a more favorable differential block than bupivacaine, the lack of equivalent clinical potency adds complexity to such comparisons.

EMLA The term eutectic is applied to mixtures in which the combination of elements has a lower melting temperature than its component elements. Lidocaine and prilocaine can combine to form such a mixture, which is marketed as EMLA (Eutectic Mixture of Local Anesthetics). This formulation, containing 2.5% of lidocaine and 2.5% prilocaine, permits anesthetic penetration of the keratinized layer of skin, producing localized numbness. It is commonly used in pediatrics to anesthetize the skin prior to venipuncture for intravenous catheter placement.

FUTURE DEVELOPMENTS Sustained-Release Formulations The provision of prolonged analgesia or anesthesia, as in the case of postoperative pain management, has traditionally been accomplished by placement of a catheter to permit continuous administration of anesthetic. More recently, efforts have focused on drug delivery systems that can slowly release anesthetic, thereby providing extended duration without the drawbacks of a catheter. Sustained-release delivery has the potential added advantage of reducing risk of systemic toxicity. Preliminary work encapsulating local anesthetic into microspheres, liposomes, and other microparticles has established proof of concept, although significant developmental problems, as well as questions regarding potential tissue toxicity, remain to be resolved.

Less Toxic Agents; More Selective Agents It has been clearly demonstrated that anesthetic neurotoxicity does not result from blockade of the voltage-gated sodium channel. Thus, effect and tissue toxicity are not mediated by a common mechanism, establishing the possibility of developing compounds with considerably better therapeutic indexes. As previously discussed, the identification and subclassification of families of neuronal sodium channels has spurred research aimed at development of more selective sodium channel blockers. The variable neuronal distribution of these isoforms and the unique role that some play in pain signaling suggests that selective blockade of these channels is feasible, and may greatly improve the therapeutic index of sodium channel modulators.

SUMMARY Drugs Used for Local Anesthesia


PREPARATIONS AVAILABLE


REFERENCES


Adverse Reactions with Bupivacaine. FDA Drug Bull 1983;13:23. Albright GA: Cardiac arrest following regional anesthesia with etidocaine or bupivacaine. Anesthesiology 1979;51:285. American Society of Regional Anesthesia and Pain Medicine: Checklist for treatment of local anesthetic systemic toxicity. 2012. http://www.asra.com/checklist-for-localanesthetic-toxicity-treatment-1-18-12.pdf. Andavan GS, Lemmens-Gruber R: Voltage-gated sodium channels: Mutations, channelopathies and targets. Curr Med Chem 2011;18:377. Auroy Y et al: Serious complications related to regional anesthesia: Results of a prospective survey in France. Anesthesiology 1997;87:479. Butterworth JF 4th, Strichartz GR: Molecular mechanisms of local anesthesia: A review. Anesthesiology 1990;72:711. Catterall WA, Goldin AL, Waxman SG: International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels. Pharmacol Rev 2005;57:397. Cave G, Harvey M: Intravenous lipid emulsion as antidote beyond local anesthetic toxicity: A systematic review. Acad Emerg Med 2009;16:815. de Jong RH, Ronfeld RA, DeRosa RA: Cardiovascular effects of convulsant and supraconvulsant doses of amide local anesthetics. Anesth Analg 1982;61:3. Di Gregorio G et al: Clinical presentation of local anesthetic systemic toxicity: A review of published cases, 1979 to 2009. Reg Anesth Pain Med 2010;35:181. Drasner K: Chloroprocaine spinal anesthesia: Back to the future? Anesth Analg 2005;100:549. Drasner K: Lidocaine spinal anesthesia: A vanishing therapeutic index? Anesthesiology 1997;87:469. Drasner K: Local anesthetic neurotoxicity: Clinical injury and strategies that may minimize risk. Reg Anesth Pain Med 2002;27:576. Drasner K: Local anesthetic systemic toxicity: a historical perspective. Reg Anesth Pain Med 2010;35:162. Drasner K et al: Cauda equina syndrome following intended epidural anesthesia. Anesthesiology 1992;77:582. Drasner K et al: Persistent sacral sensory deficit induced by intrathecal local anesthetic infusion in the rat. Anesthesiology 1994;80:847. Freedman JM et al: T ransient neurologic symptoms after spinal anesthesia: An epidemiologic study of 1,863 patients. Anesthesiology 1998;89:633. Goldblum E, Atchabahian A: T he use of 2-chloroprocaine for spinal anaesthesia. Acta Anaesthesiol Scand 2013;57:545. Groban L: Central nervous system and cardiac effects from long-acting amide local anesthetic toxicity in the intact animal model. Reg Anesth Pain Med 2003;28:3. Hampl KF et al: T ransient neurologic symptoms after spinal anesthesia. Anesth Analg 1995;81:1148. Hille B: Local anesthetics: Hydrophyilic and hydrophobic pathways for the drug-receptor interaction. J Gen Physiol 1977;69:497. Holmdahl MH: Xylocain (lidocaine, lignocaine), its discovery and Gordh’s contribution to its clinical use. Acta Anaesthesiol Scand Suppl 1998;113:8. Kouri ME, Kopacz DJ: Spinal 2-chloroprocaine: A comparison with lidocaine in volunteers. Anesth Analg 2004;98(1):75. Kuo I, Akpa BS: Validity of the lipid sink as a mechanism for the reversal of local anesthetic systemic toxicity: A physiologically based pharmacokinetic model study. Anesthesiology 2013;118:1350. Mattison JB: Cocaine poisoning. Med Surg Rep 1891;115:645. Neal JM et al: ASRA practice advisory on local anesthetic systemic toxicity. Reg Anesth Pain Med 2010;35:152. Pollock JE: T ransient neurologic symptoms: Etiology, risk factors, and management. Reg Anesth Pain Med 2002;27:581. Pollock JE et al: Prospective study of the incidence of transient radicular irritation in patients undergoing spinal anesthesia. Anesthesiology 1996;84:1361. Priest BT : Future potential and status of selective sodium channel blockers for the treatment of pain. Curr Opin Drug Discov Devel 2009;12:682. Rigler ML et al: Cauda equina syndrome after continuous spinal anesthesia. Anesth Analg 1991;72:275. Rose JS, Neal JM, Kopacz DJ: Extended-duration analgesia: Update on microspheres and liposomes. Reg Anesth Pain Med 2005;30:275. Ruetsch YA, Boni T , Borgeat A: From cocaine to ropivacaine: T he history of local anesthetic drugs. Curr T op Med Chem 2001;1:175. Sakura S et al: Local anesthetic neurotoxicity does not result from blockade of voltage-gated sodium channels. Anesth Analg 1995;81:338. Schneider M et al: T ransient neurologic toxicity after hyperbaric subarachnoid anesthesia with 5% lidocaine. Anesth Analg 1993;76:1154. Sirianni AJ et al: Use of lipid emulsion in the resuscitation of a patient with prolonged cardiovascular collapse after overdose of bupropion and lamotrigine. Ann Emerg Med 2008;51:412. T aniguchi M, Bollen AW, Drasner K: Sodium bisulfite: Scapegoat for chloroprocaine neurotoxicity? Anesthesiology 2004;100:85. T remont-Lukats IW et al: Systemic administration of local anesthetics to relieve neuropathic pain: A systematic review and meta-analysis. Anesth Analg 2005;101:1738. Weinberg GL: Lipid resuscitation: More than a sink. Crit Care Med 2012;40:2521. Weinberg GL et al: Pretreatment or resuscitation with a lipid infusion shifts the dose-response to bupivacaine-induced asystole in rats. Anesthesiology 1998;88:1071. *

The author thanks Bertram G. Katzung, MD, PhD, and Paul F. White, PhD, MD, for contributions to this chapter in previous editions.

CASE STUDY ANSWER If a spinal anesthetic technique were selected, bupivacaine would be an excellent choice. It has an adequately long duration of action and a relatively unblemished record with respect to neurotoxic injury and transient neurologic symptoms, which are the complications of most concern with spinal anesthetic technique. Although bupivacaine has greater potential for cardiotoxicity, this is not a concern when the drug is used for spinal anesthesia because of the extremely low doses required for intrathecal administration. If an epidural technique were chosen for the surgical procedure, the potential for systemic toxicity would need to be considered, making lidocaine or mepivacaine (generally with epinephrine) preferable to bupivacaine (or even ropivacaine or levobupivacaine) because of their better therapeutic indexes with respect to cardiotoxicity. However, this does not apply to epidural administration for postoperative pain control, which involves administration of more dilute anesthetic at a slower rate. The most common agents used for this indication are bupivacaine, ropivacaine, and levobupivacaine.


CHAPTER

27 Skeletal Muscle Relaxants Marieke Kruidering-Hall, PhD, & Lundy Campbell, MD*

CASE STUDY A 30-year-old woman is rushed to the emergency department at a major trauma center after a motor vehicle crash. Although in significant pain, she is awake, alert, and able to give a brief history. She states that she was the driver and was wearing a seatbelt. There were no passengers in the car. Her past medical history is significant only for asthma, for which she has been intubated once in the past. She has no allergies to medications. There are multiple lacerations on her face and extremities and a large open fracture of her right femur. An orthopedic surgeon has scheduled immediate operative repair of the femur fracture, and the plastic surgeon wants to suture the facial lacerations at the same time. You decide to intubate the patient for the procedure. What muscle relaxant would you choose? Would you choose the same agent if she had experienced a 30% total body burn in a fire at the time of the accident? What if the past medical history included right-sided hemiparesis of 10 years’ duration?

Drugs that affect skeletal muscle function include two different therapeutic groups: those used during surgical procedures and in the intensive care unit (ICU) to produce muscle paralysis (ie, neuromuscular blockers), and those used to reduce spasticity in a variety of painful conditions (ie, spasmolytics). Neuromuscular blocking drugs interfere with transmission at the neuromuscular end plate and lack central nervous system (CNS) activity. These compounds are used primarily as adjuncts during general anesthesia to optimize surgical conditions and to facilitate endotracheal intubation in order to ensure adequate ventilation. Drugs in the spasmolytic group have traditionally been called “centrally acting” muscle relaxants and are used primarily to treat chronic back pain and painful fibromyalgic conditions. Dantrolene, a spasmolytic agent that has no significant central effects and is used primarily to treat a rare anesthetic-related complication, malignant hyperthermia, is also discussed in this chapter.

NEUROMUSCULAR BLOCKING DRUGS History During the 16th century, European explorers found that natives in the Amazon Basin of South America were using curare, an arrow poison that produced skeletal muscle paralysis, to kill animals. The active compound, d-tubocurarine, and its modern synthetic analogs have had a major influence on the practice of anesthesia and surgery and have proved useful in understanding the basic mechanisms involved in neuromuscular transmission.

Normal Neuromuscular Function The mechanism of neuromuscular transmission at the motor end plate is similar to that described for preganglionic cholinergic nerves in Chapter 6. The arrival of an action potential at the motor nerve terminal causes an influx of calcium and release of the neurotransmitter acetylcholine. Acetylcholine then diffuses across the synaptic cleft to activate nicotinic receptors located on the motor end plate, present at a density of 10,000/μm* . As noted in Chapter 7, the adult NM receptor is composed of five peptides: two alpha peptides, one beta, one gamma, and one delta peptide (Figure 27–1). The binding of two acetylcholine molecules to receptors on the α-β and δ-α subunits causes opening of the channel. The subsequent movement of sodium and potassium through the channel is associated with a graded depolarization of the end plate membrane (see Figure 7–4, panel B). This change in voltage is termed the motor end plate potential. The


magnitude of the end plate potential is directly related to the amount of acetylcholine released. If the potential is small, the permeability and the end plate potential return to normal without an impulse being propagated from the end plate region to the rest of the muscle membrane. However, if the end plate potential is large, the adjacent muscle membrane is depolarized, and an action potential will be propagated along the entire muscle fiber. Muscle contraction is then initiated by excitation-contraction coupling. The released acetylcholine is quickly removed from the end plate region by both diffusion and enzymatic destruction by the local acetylcholinesterase enzyme.


FIGURE 27–1 The adult nicotinic acetylcholine receptor (nAChR) is an intrinsic membrane protein with five distinct subunits (α2 β δ γ). A: Cartoon of the one of five subunits of the AChR in the end plate surface of adult mammalian muscle. Each subunit contains four


helical domains labeled M1 to M4. The M2 domains line the channel pore. B: Cartoon of the full nAChR. The N termini of two subunits cooperate to form two distinct binding pockets for acetylcholine (ACh). These pockets occur at the α-β and the δ-α subunit interfaces. Binding of one molecule of ACh enhances the receptor’s affinity for the second molecule, followed by multiple intermediate steps leading to channel opening. These steps are the subject of intense investigation. At least two additional types of acetylcholine receptors are found within the neuromuscular apparatus. One type is located on the presynaptic motor axon terminal, and activation of these receptors mobilizes additional transmitter for subsequent release by moving more acetylcholine vesicles toward the synaptic membrane. The second type of receptor is found on extrajunctional cells and is not normally involved in neuromuscular transmission. However, under certain conditions (eg, prolonged immobilization, thermal burns), these receptors may proliferate sufficiently to affect subsequent neuromuscular transmission. This proliferation of extrajunctional acetylcholine receptors may be clinically relevant when using depolarizing or nondepolarizing skeletal muscle relaxant drugs and is described later. Skeletal muscle relaxation and paralysis can occur from interruption of function at several sites along the pathway from the CNS to myelinated somatic nerves, unmyelinated motor nerve terminals, nicotinic acetylcholine receptors, the motor end plate, the muscle membrane, and the intracellular muscular contractile apparatus itself. Blockade of end plate function can be accomplished by two basic mechanisms. First, pharmacologic blockade of the physiologic agonist acetylcholine is characteristic of the antagonist neuromuscular blocking drugs (ie, nondepolarizing neuromuscular blocking drugs). These drugs prevent access of the transmitter to its receptor and thereby prevent depolarization. The prototype of this nondepolarizing subgroup is d-tubocurarine. The second mechanism of blockade can be produced by an excess of a depolarizing agonist, such as acetylcholine. This seemingly paradoxical effect of acetylcholine also occurs at the ganglionic nicotinic acetylcholine receptor. The prototypical depolarizing blocking drug is succinylcholine. A similar depolarizing block can be produced by acetylcholine itself when high local concentrations are achieved in the synaptic cleft (eg, by cholinesterase inhibitor intoxication) and by nicotine and other nicotinic agonists. However, the neuromuscular block produced by depolarizing drugs other than succinylcholine cannot be precisely controlled and is of no clinical value.

BASIC PHARMACOLOGY OF NEUROMUSCULAR BLOCKING DRUGS Chemistry All of the available neuromuscular blocking drugs bear a structural resemblance to acetylcholine. For example, succinylcholine is two acetylcholine molecules linked end-to-end (Figure 27–2). In contrast to the single linear structure of succinylcholine and other depolarizing drugs, the nondepolarizing agents (eg, pancuronium) conceal the “double-acetylcholine” structure in one of two types of bulky, semirigid ring systems (Figure 27–2). Examples of the two major families of nondepolarizing blocking drugs—the isoquinoline and steroid derivatives—are shown in Figures 27–3 and 27–4. Another feature common to all currently used neuromuscular blockers is the presence of one or two quaternary nitrogens, which makes them poorly lipid soluble and limits entry into the CNS.


FIGURE 27–2 Structural relationship of succinylcholine, a depolarizing agent, and pancuronium, a nondepolarizing agent, to acetylcholine, the neuromuscular transmitter. Succinylcholine, originally called diacetylcholine, is simply two molecules of acetylcholine linked through the acetate methyl groups. Pancuronium may be viewed as two acetylcholine-like fragments (outlined in color) oriented on a steroid nucleus.


FIGURE 27–3 Structures of two isoquinoline neuromuscular blocking drugs. These agents are nondepolarizing muscle relaxants.



FIGURE 27–4 Structures of steroid neuromuscular blocking drugs (steroid nucleus in color). These agents are all nondepolarizing muscle relaxants.

Pharmacokinetics of Neuromuscular Blocking Drugs All of the neuromuscular blocking drugs are highly polar compounds and inactive orally; they must be administered parenterally. A. Nondepolarizing Relaxant Drugs The rate of disappearance of a nondepolarizing neuromuscular blocking drug from the blood is characterized by a rapid initial distribution phase followed by a slower elimination phase. Neuromuscular blocking drugs are highly ionized, do not readily cross cell membranes, and are not strongly bound in peripheral tissues. Therefore, their volume of distribution (80–140 mL/kg) is only slightly larger than the blood volume. The duration of neuromuscular blockade produced by nondepolarizing relaxants is strongly correlated with the elimination half-life. Drugs that are excreted by the kidney typically have longer half-lives, leading to longer durations of action (> 35 minutes). Drugs eliminated by the liver tend to have shorter half-lives and durations of action (Table 27–1). All steroidal muscle relaxants are metabolized to their 3-hydroxy, 17-hydroxy, or 3,17-dihydroxy products in the liver. The 3-hydroxy metabolites are usually 40–80% as potent as the parent drug. Under normal circumstances, metabolites are not formed in sufficient quantities to produce a significant degree of neuromuscular blockade during or after anesthesia. However, if the parent compound is administered for several days in the ICU setting, the 3-hydroxy metabolite may accumulate and cause prolonged paralysis because it has a longer half-life than the parent compound. The remaining metabolites possess minimal neuromuscular blocking properties. TABLE 27–1 Pharmacokinetic and dynamic properties of neuromuscular blocking drugs.

The intermediate-acting steroid muscle relaxants (eg, vecuronium and rocuronium) tend to be more dependent on biliary excretion


or hepatic metabolism for their elimination. These muscle relaxants are more commonly used clinically than the long-acting steroid-based drugs (eg, pancuronium). The duration of action of these relaxants may be prolonged significantly in patients with impaired liver function. Atracurium (Figure 27–3) is an intermediate-acting isoquinoline nondepolarizing muscle relaxant that is no longer in widespread clinical use. In addition to hepatic metabolism, atracurium is inactivated by a form of spontaneous breakdown known as Hofmann elimination. The main breakdown products are laudanosine and a related quaternary acid, neither of which possesses neuromuscular blocking properties. Laudanosine is slowly metabolized by the liver and has a longer elimination half-life (ie, 150 minutes). It readily crosses the blood-brain barrier, and high blood concentrations may cause seizures and an increase in the volatile anesthetic requirement. During surgical anesthesia, blood levels of laudanosine typically range from 0.2 to 1 mcg/mL; however, with prolonged infusions of atracurium in the ICU, laudanosine blood levels may exceed 5 mcg/mL. Atracurium has several stereoisomers, and the potent isomer cisatracurium has become one of the most common muscle relaxants in use today. Although cisatracurium resembles atracurium, it has less dependence on hepatic inactivation, produces less laudanosine, and is much less likely to release histamine. From a clinical perspective, cisatracurium has all the advantages of atracurium with fewer adverse effects. Therefore, cisatracurium has virtually replaced atracurium in clinical practice. Gantacurium represents a new class of nondepolarizing neuromuscular blockers, called asymmetric mixed-onium chlorofumarates. It is degraded nonenzymatically by adduction of the amino acid cysteine and ester bond hydrolysis. Gantacurium is currently in phase 3 clinical trials and not yet available for widespread clinical use. Preclinical and clinical data indicate gantacurium has a rapid onset of effect and predictable duration of action (very short, similar to succinylcholine) that can be reversed with neostigmine or more quickly (within 1–2 minutes), with administration of L-cysteine. At doses above three times the ED 95 , cardiovascular adverse effects (eg, hypotension) have occurred, probably due to histamine release. No bronchospasm or pulmonary vasoconstriction has been reported at these higher doses. B. Depolarizing Relaxant Drugs The extremely short duration of action of succinylcholine (5–10 minutes) is due to its rapid hydrolysis by butyrylcholinesterase and pseudocholinesterase in the liver and plasma, respectively. Plasma cholinesterase metabolism is the predominant pathway for succinylcholine elimination. The primary metabolite of succinylcholine, succinylmonocholine, is rapidly broken down to succinic acid and choline. Because plasma cholinesterase has an enormous capacity to hydrolyze succinylcholine, only a small percentage of the original intravenous dose ever reaches the neuromuscular junction. In addition, because there is little if any plasma cholinesterase at the motor end plate, a succinylcholine-induced blockade is terminated by its diffusion away from the end plate into extracellular fluid. Therefore, the circulating levels of plasma cholinesterase influence the duration of action of succinylcholine by determining the amount of the drug that reaches the motor end plate. Neuromuscular blockade produced by succinylcholine can be prolonged in patients with an abnormal genetic variant of plasma cholinesterase. The dibucaine number is a measure of the ability of a patient to metabolize succinylcholine and can be used to identify at-risk patients. Under standardized test conditions, dibucaine inhibits the normal enzyme by 80% and the abnormal enzyme by only 20%. Many genetic variants of plasma cholinesterase have been identified, although the dibucaine-related variants are the most important. Given the rarity of these genetic variants, plasma cholinesterase testing is not a routine clinical procedure but may be indicated for patients with a family history of plasma cholinesterase deficiency. Another reasonable strategy is to avoid the use of succinylcholine where practical in patients with a possible family history of plasma cholinesterase deficiency.

Mechanism of Action The interactions of drugs with the acetylcholine receptor-end plate channel have been described at the molecular level. Several modes of action of drugs on the receptor are illustrated in Figure 27–5.


FIGURE 27–5 Schematic diagram of the interactions of drugs with the acetylcholine receptor on the end plate channel (structures are purely symbolic). Top: The action of the normal agonist, acetylcholine (red) in opening the channel. Bottom, left: A nondepolarizing blocker, eg, rocuronium (yellow), is shown as preventing the opening of the channel when it binds to the receptor. Bottom, right: A depolarizing blocker, eg, succinylcholine (blue), both occupying the receptor and blocking the channel. Normal closure of the channel gate is prevented and the blocker may move rapidly in and out of the pore. Depolarizing blockers may desensitize the end plate by occupying the receptor and causing persistent depolarization. An additional effect of drugs on the end plate channel may occur through changes in the lipid environment surrounding the channel (not shown). General anesthetics and alcohols may impair neuromuscular transmission by this mechanism. A. Nondepolarizing Relaxant Drugs All the neuromuscular blocking drugs in current use in the USA except succinylcholine are classified as nondepolarizing agents. Although it is no longer in widespread clinical use, d-tubocurarine is considered the prototype neuromuscular blocker. When small doses of nondepolarizing muscle relaxants are administered, they act predominantly at the nicotinic receptor site by competing with acetylcholine. The least potent nondepolarizing relaxants (eg, rocuronium) have the fastest onset and the shortest duration of action. In larger doses, nondepolarizing drugs can enter the pore of the ion channel (Figure 27–1) to produce a more intense motor blockade. This action further weakens neuromuscular transmission and diminishes the ability of the acetylcholinesterase inhibitors (eg, neostigmine, edrophonium, pyridostigmine) to antagonize the effect of nondepolarizing muscle relaxants. Nondepolarizing relaxants can also block prejunctional sodium channels. As a result of this action, muscle relaxants interfere with the mobilization of acetylcholine at the nerve ending and cause fade of evoked nerve twitch contractions (Figure 27–6, and described below). One consequence of the surmountable nature of the postsynaptic blockade produced by nondepolarizing muscle relaxants is the fact that


tetanic stimulation (rapid delivery of electrical stimuli to a peripheral nerve) releases a large quantity of acetylcholine and is followed by transient posttetanic facilitation of the twitch strength (ie, relief of blockade). An important clinical consequence of this principle is the reversal of residual blockade by cholinesterase inhibitors. The characteristics of a nondepolarizing neuromuscular blockade are summarized in Table 27–2 and Figure 27–6.

FIGURE 27–6 Muscle contraction responses to different patterns of nerve stimulation used in monitoring skeletal muscle relaxation. The alterations produced by a nondepolarizing blocker and depolarizing and desensitizing blockade by succinylcholine are shown. In the train-of-four (TOF) pattern, four stimuli are applied at 2 Hz. The TOF ratio (TOF-R) is calculated from the strength of the fourth contraction divided by that of the first. In the double-burst pattern, three stimuli are applied at 50 Hz, followed by a 700 ms rest period and then repeated. In the posttetanic potentiation pattern, several seconds of 50 Hz stimulation are applied, followed by several seconds of rest and then by single stimuli at a slow rate (eg, 0.5 Hz). The number of detectable posttetanic twitches is the posttetanic count (PTC).* , first posttetanic contraction. TABLE 27–2 Comparison of a typical nondepolarizing muscle relaxant (rocuronium) and a depolarizing muscle relaxant (succinylcholine).


B. Depolarizing Relaxant Drugs 1. Phase I block (depolarizing)—Succinylcholine is the only clinically useful depolarizing blocking drug. Its neuromuscular effects are like those of acetylcholine except that succinylcholine produces a longer effect at the myoneural junction. Succinylcholine reacts with the nicotinic receptor to open the channel and cause depolarization of the motor end plate, and this in turn spreads to the adjacent membranes, causing contractions of muscle motor units. Data from single-channel recordings indicate that depolarizing blockers can enter the channel to produce a prolonged “flickering” of the ion conductance (Figure 27–7). Because succinylcholine is not metabolized effectively at the synapse, the depolarized membranes remain depolarized and unresponsive to subsequent impulses (ie, a state of depolarizing blockade). Furthermore, because excitation-contraction coupling requires end plate repolarization (“repriming”) and repetitive firing to maintain muscle tension, a flaccid paralysis results. In contrast to the nondepolarizing drugs, this so-called phase I (depolarizing) block is augmented, not reversed, by cholinesterase inhibitors.


FIGURE 27–7 Action of succinylcholine on single-channel end plate receptor currents in frog muscle. Currents through a single AChR channel were recorded using the patch clamp technique. The upper trace was recorded in the presence of a low concentration of succinylcholine; the downward deflections represent openings of the channel and passage of inward (depolarizing) current. The lower trace was recorded in the presence of a much higher concentration of succinylcholine and shows prolonged “flickering” of the channel as it repetitively opens and closes or is “plugged” by the drug. (Reproduced, with permission, from Marshall CG, Ogden DC, Colquhoun D: The actions of suxamethonium (succinyldicholine) as an agonist and channel blocker at the nicotinic receptor of frog muscle. J Physiol [Lond] 1990;428:155.) The characteristics of a depolarizing neuromuscular blockade are summarized in Table 27–2 and Figure 27–6. 2. Phase II block (desensitizing)—With prolonged exposure to succinylcholine, the initial end plate depolarization decreases and the membrane becomes repolarized. Despite this repolarization, the membrane cannot easily be depolarized again because it is desensitized. The mechanism for this desensitizing phase is unclear, but some evidence indicates that channel block may become more important than agonist action at the receptor in phase II of succinylcholine’s neuromuscular blocking action. Regardless of the mechanism, the channels behave as if they are in a prolonged closed state (Figure 27–6). Later in phase II, the characteristics of the blockade are nearly identical to those of a nondepolarizing block (ie, a nonsustained twitch response to a tetanic stimulus) (Figure 27–6), with possible reversal by acetylcholinesterase inhibitors.

CLINICAL PHARMACOLOGY OF NEUROMUSCULAR BLOCKING DRUGS Skeletal Muscle Paralysis Before the introduction of neuromuscular blocking drugs, profound skeletal muscle relaxation for intracavitary operations could be achieved only by producing levels of volatile (inhaled) anesthesia deep enough to produce profound depressant effects on the cardiovascular and respiratory systems. The adjunctive use of neuromuscular blocking drugs makes it possible to achieve adequate muscle relaxation for all types of surgical procedures without the cardiorespiratory depressant effects produced by deep anesthesia.

Assessment of Neuromuscular Transmission Monitoring the effect of muscle relaxants during surgery (and recovery following the administration of cholinesterase inhibitors) typically involves the use of a device that produces transdermal electrical stimulation of one of the peripheral nerves to the hand or facial muscles and recording of the evoked contractions (ie, twitch responses). The motor responses to different patterns of peripheral nerve stimulation can be recorded in the operating room during the procedure (Figure 27–6). The standard approach for monitoring the clinical effects of muscle relaxants during surgery uses peripheral nerve stimulation to elicit motor responses, which are visually observed by the anesthesiologist. The three most commonly used patterns include (1) single-twitch stimulation, (2) train-of-four (TOF) stimulation, and (3)


tetanic stimulation. Two other modalities are also available to monitor neuromuscular transmission: double-burst stimulation and posttetanic count. With single-twitch stimulation, a single supramaximal electrical stimulus is applied to a peripheral nerve at frequencies from 0.1 Hz to 1.0 Hz. The higher frequency is often used during induction and reversal to more accurately determine the peak (maximal) drug effect. TOF stimulation involves four successive supramaximal stimuli given at intervals of 0.5 second (2 Hz). Each stimulus in the TOF causes the muscle to contract, and the relative magnitude of the response of the fourth twitch compared with the first twitch is the TOF ratio. With a depolarizing block, all four twitches are reduced in a dose-related fashion. With a nondepolarizing block, the TOF ratio decreases (“fades”) and is inversely proportional to the degree of blockade. During recovery from nondepolarizing block, the amount of fade decreases and the TOF ratio approaches 1.0. Recovery to a TOF ratio greater than 0.7 is typically necessary for resumption of spontaneous ventilation. However, complete clinical recovery from a nondepolarizing block is considered to require a TOF greater than 0.9. Fade in the TOF response after administration of succinylcholine signifies the development of a phase II block. Tetanic stimulation consists of a very rapid (30–100 Hz) delivery of electrical stimuli for several seconds. During a nondepolarizing neuromuscular block (and a phase II block after succinylcholine), the response is not sustained and fade of the twitch responses is observed. Fade in response to tetanic stimulation is normally considered a presynaptic event. However, the degree of fade depends primarily on the degree of neuromuscular blockade. During a partial nondepolarizing blockade, tetanic nerve stimulation is followed by an increase in the posttetanic twitch response, so-called posttetanic facilitation of neuromuscular transmission. During intense neuromuscular blockade, there is no response to either tetanic or posttetanic stimulation. As the intensity of the block diminishes, the response to posttetanic twitch stimulation reappears. The reappearance of the first response to twitch stimulation after tetanic stimulation reflects the duration of profound (clinical) neuromuscular blockade. To determine the posttetanic count, 5 seconds of 50 Hz tetany is applied, followed by 3 seconds of rest, followed by 1 Hz pulses for about 10 seconds (10 pulses). The counted number of muscle twitches provides an estimation of the depth of blockade. For instance, a posttetanic count of 2 suggests no twitch response (by TOF) for about 20–30 minutes, and a posttetanic count of 5 correlates to a no-twitch response (by TOF) of about 10–15 minutes (Figure 27–6, bottom panel). The double-burst stimulation pattern is another mode of electrical nerve stimulation developed with the goal of allowing for manual detection of residual neuromuscular blockade when it is not possible to record the responses to single-twitch, TOF, or tetanic stimulation. In this pattern, three nerve stimuli are delivered at 50 Hz followed by a 700 ms rest period and then by two or three additional stimuli at 50 Hz. It is easier to detect fade in the responses to double-burst stimulation than to TOF stimulation. The absence of fade in response to double-burst stimulation implies that clinically significant residual neuromuscular blockade does not exist. A more quantitative approach to neuromuscular monitoring involves monitoring using a force transducer for measuring the evoked response (ie, movement) of the thumb to TOF stimulation over the ulnar nerve at the wrist. This device has the advantage of being integrated in the anesthesia machine and also provides a more accurate graphic display of the percentage of fade to TOF stimulation. A. Nondepolarizing Relaxant Drugs During anesthesia, administration of tubocurarine, 0.1–0.4 mg/kg IV, initially causes motor weakness, followed by the skeletal muscles becoming flaccid and inexcitable to electrical stimulation (Figure 27–8). In general, larger muscles (eg, abdominal, trunk, paraspinous, diaphragm) are more resistant to neuromuscular blockade and recover more rapidly than smaller muscles (eg, facial, foot, hand). The diaphragm is usually the last muscle to be paralyzed. Assuming that ventilation is adequately maintained, no adverse effects occur with skeletal muscle paralysis. When administration of muscle relaxants is discontinued, recovery of muscles usually occurs in reverse order, with the diaphragm regaining function first. The pharmacologic effect of tubocurarine, 0.3 mg/kg IV, usually lasts 45–60 minutes. However, subtle evidence of residual muscle paralysis detected using a neuromuscular monitor may last for another hour, increasing the likelihood of adverse outcomes, eg, aspiration and decreased hypoxic drive. Potency and duration of action of the other nondepolarizing drugs are shown in Table 27–1. In addition to the duration of action, the most important property distinguishing the nondepolarizing relaxants is the time to onset of the blocking effect, which determines how rapidly the patient’s trachea can be intubated. Of the currently available nondepolarizing drugs, rocuronium has the most rapid onset time (60–120 seconds).


FIGURE 27–8 Neuromuscular blockade from tubocurarine during equivalent levels of isoflurane and halothane anesthesia in patients. Note that isoflurane augments the block far more than does halothane. MAC, minimal alveolar concentration. B. Depolarizing Relaxant Drugs Following the administration of succinylcholine, 0.75–1.5 mg/kg IV, transient muscle fasciculations occur over the chest and abdomen within 30 seconds, although general anesthesia and the prior administration of a small dose of a nondepolarizing muscle relaxant tends to attenuate them. As paralysis develops rapidly (< 90 seconds), the arm, neck, and leg muscles are initially relaxed followed by the respiratory muscles. As a result of succinylcholine’s rapid hydrolysis by cholinesterase in the plasma (and liver), the duration of neuromuscular block typically lasts less than 10 minutes (Table 27–1).

Cardiovascular Effects Vecuronium, cisatracurium, and rocuronium have minimal, if any, cardiovascular effects. The other nondepolarizing muscle relaxants (ie, pancuronium and atracurium) produce cardiovascular effects that are mediated by autonomic or histamine receptors (Table 27–3). Tubocurarine and, to a lesser extent, atracurium can produce hypotension as a result of systemic histamine release, and with larger doses, ganglionic blockade may occur with tubocurarine. Premedication with an antihistaminic compound attenuates tubocurarine-induced hypotension. Pancuronium causes a moderate increase in heart rate and a smaller increase in cardiac output, with little or no change in systemic vascular resistance. Although pancuronium-induced tachycardia is primarily due to a vagolytic action, release of norepinephrine from adrenergic nerve endings and blockade of neuronal uptake of norepinephrine may be secondary mechanisms. Bronchospasm may be produced by neuromuscular blockers that release histamine (eg, atracurium), but after induction of general anesthesia, insertion of an endotracheal tube is the most common cause of bronchospasm. TABLE 27–3 Effects of neuromuscular blocking drugs on other tissues.


Succinylcholine can cause cardiac arrhythmias, especially when administered during halothane anesthesia. The drug stimulates autonomic cholinoceptors, including the nicotinic receptors at both sympathetic and parasympathetic ganglia and muscarinic receptors in the heart (eg, sinus node). The negative inotropic and chronotropic responses to succinylcholine can be attenuated by administration of an anticholinergic drug (eg, glycopyrrolate, atropine). With large doses of succinylcholine, positive inotropic and chronotropic effects may be observed. On the other hand, bradycardia has been repeatedly observed when a second dose of succinylcholine is given less than 5 minutes after the initial dose. This transient bradycardia can be prevented by thiopental, atropine, ganglionic-blocking drugs, and by pretreating with a small dose of a nondepolarizing muscle relaxant (eg, rocuronium). Direct myocardial effects, increased muscarinic stimulation, and ganglionic stimulation contribute to this bradycardic response.

Other Adverse Effects of Depolarizing Blockade A. Hyperkalemia Patients with burns, nerve damage or neuromuscular disease, closed head injury, and other trauma may develop proliferation of extrajunctional acetylcholine receptors. During administration of succinylcholine, potassium is released from muscles, likely due to fasciculations. If the proliferation of extrajunctional receptors is great enough, sufficient potassium may be released to result in cardiac arrest. The exact time course of receptor proliferation is unknown; therefore, it is best to avoid the use of succinylcholine in these cases. B. Increased Intraocular Pressure Administration of succinylcholine may be associated with the rapid onset of an increase in intraocular pressure (< 60 seconds), peaking at 2–4 minutes, and declining after 5 minutes. The mechanism may involve tonic contraction of myofibrils or transient dilation of ocular choroidal blood vessels. Despite the increase in intraocular pressure, the use of succinylcholine for ophthalmologic operations is not contraindicated unless the anterior chamber is open (“open globe�) due to trauma.


C. Increased Intragastric Pressure In heavily muscled patients, the fasciculations associated with succinylcholine may cause an increase in intragastric pressure ranging from 5 to 40 cm H2 O, increasing the risk for regurgitation and aspiration of gastric contents. This complication is more likely to occur in patients with delayed gastric emptying (eg, those with diabetes), traumatic injury (eg, an emergency case), esophageal dysfunction, and morbid obesity. D. Muscle Pain Myalgias are a common postoperative complaint of heavily muscled patients and those who receive large doses (> 1.5 mg/kg) of succinylcholine. The true incidence of myalgias related to muscle fasciculations is difficult to establish because of confounding factors, including the anesthetic technique, type of surgery, and positioning during the operation. However, the incidence of myalgias has been reported to vary from less than 1% to 20%. It occurs more frequently in ambulatory than in bedridden patients. The pain is thought to be secondary to the unsynchronized contractions of adjacent muscle fibers just before the onset of paralysis. However, there is controversy over whether the incidence of muscle pain following succinylcholine is actually higher than with nondepolarizing muscle relaxants when other potentially confounding factors are taken into consideration.

Interactions with Other Drugs A. Anesthetics Inhaled (volatile) anesthetics potentiate the neuromuscular blockade produced by nondepolarizing muscle relaxants in a dose-dependent fashion. Of the general anesthetics that have been studied, inhaled anesthetics augment the effects of muscle relaxants in the following order: isoflurane (most); sevoflurane, desflurane, halothane; and nitrous oxide (least) (Figure 27–8). The most important factors involved in this interaction are the following: (1) nervous system depression at sites proximal to the neuromuscular junction (ie, CNS); (2) increased muscle blood flow (ie, due to peripheral vasodilation produced by volatile anesthetics), which allows a larger fraction of the injected muscle relaxant to reach the neuromuscular junction; and (3) decreased sensitivity of the postjunctional membrane to depolarization. A rare interaction of succinylcholine with volatile anesthetics results in malignant hyperthermia, a condition caused by abnormal release of calcium from stores in skeletal muscle. This condition is treated with dantrolene and is discussed below under Spasmolytic Drugs and in Chapter 16. B. Antibiotics Numerous reports have described enhancement of neuromuscular blockade by antibiotics (eg, aminoglycosides). Many of the antibiotics have been shown to cause a depression of evoked release of acetylcholine similar to that caused by administering magnesium. The mechanism of this prejunctional effect appears to be blockade of specific P-type calcium channels in the motor nerve terminal. C. Local Anesthetics and Antiarrhythmic Drugs In small doses, local anesthetics can depress posttetanic potentiation via a prejunctional neural effect. In large doses, local anesthetics can block neuromuscular transmission. With these higher doses, local anesthetics block acetylcholine-induced muscle contractions as a result of blockade of the nicotinic receptor ion channels. Experimentally, similar effects can be demonstrated with sodium channelblocking antiarrhythmic drugs such as quinidine. However, at the doses used for cardiac arrhythmias, this interaction is of little or no clinical significance. Higher doses of bupivacaine have been associated with cardiac arrhythmias independent of the muscle relaxant used. D. Other Neuromuscular Blocking Drugs The end plate-depolarizing effect of succinylcholine can be antagonized by administering a small dose of a nondepolarizing blocker. To prevent the fasciculations associated with succinylcholine administration, a small nonparalyzing dose of a nondepolarizing drug can be given before succinylcholine (eg, d-tubocurarine, 2 mg IV, or pancuronium, 0.5 mg IV). Although this dose usually reduces fasciculations and postoperative myalgias, it can increase the amount of succinylcholine required for relaxation by 50–90% and can produce a feeling of weakness in awake patients. Therefore, “pre-curarization” before succinylcholine is no longer widely practiced.

Effects of Diseases & Aging on the Neuromuscular Response Several diseases can diminish or augment the neuromuscular blockade produced by nondepolarizing muscle relaxants. Myasthenia gravis enhances the neuromuscular blockade produced by these drugs. Advanced age is associated with a prolonged duration of action from nondepolarizing relaxants as a result of decreased clearance of the drugs by the liver and kidneys. As a result, the dosage of neuromuscular blocking drugs should be reduced in older patients (> 70 years).


Conversely, patients with severe burns and those with upper motor neuron disease are resistant to nondepolarizing muscle relaxants. This desensitization is probably caused by proliferation of extrajunctional receptors, which results in an increased dose requirement for the nondepolarizing relaxant to block a sufficient number of receptors.

Reversal of Nondepolarizing Neuromuscular Blockade The cholinesterase inhibitors effectively antagonize the neuromuscular blockade caused by nondepolarizing drugs. Their general pharmacology is discussed in Chapter 7. Neostigmine and pyridostigmine antagonize nondepolarizing neuromuscular ablockade by increasing the availability of acetylcholine at the motor end plate, mainly by inhibition of acetylcholinesterase. To a lesser extent, these cholinesterase inhibitors also increase the release of this transmitter from the motor nerve terminal. In contrast, edrophonium antagonizes neuromuscular blockade purely by inhibiting acetylcholinesterase activity. Edrophonium has a more rapid onset of action but may be less effective than neostigmine in reversing the effects of nondepolarizing blockers in the presence of profound neuromuscular blockade. These differences are important in determining recovery from residual block , the neuromuscular blockade remaining after completion of surgery and movement of the patient to the recovery room. Unsuspected residual block may result in hypoventilation, leading to hypoxia and even apnea, especially if patients have received central depressant medications in the early recovery period. Sugammadex is a novel reversal agent approved in Europe. It is still in phase 3 clinical trials and not yet approved for use in the USA. Its approval has been delayed over concerns that it may induce coagulopathy and hypersensitivity reactions. Sugammadex is a modified γ-cyclodextrin (a macro-ring structure with 16 polar hydroxyl groups facing inward and 8 polar carboxyl groups facing outward) that binds tightly to rocuronium in a 1:1 ratio. By binding to plasma rocuronium, sugammadex decreases the free plasma concentration and establishes a concentration gradient for rocuronium to diffuse away from the neuromuscular junction back into the circulation, where it is quickly bound by free sugammadex. Sugammadex will bind to and can reverse effects of other steroidal neuromuscular blockers such as vecuronium and pancuronium, but to a lesser extent. Clinical trials studying the safety and efficacy of sugammadex have used doses varying between 0.5 and 16 mg/kg. These trials reported no difference in prevalence of untoward effects among sugammadex, placebo, and neostigmine. Currently, three dose ranges are recommended: 2 mg/kg to reverse shallow neuromuscular blockade, 4 mg/kg to reverse profound blockade (1–2 posttetanic count), and 1 mg/kg for immediate reversal following administration of rocuronium. The sugammadex-rocuronium complex is typically excreted unchanged in the urine within 24 hours in patients with normal renal function. In patients with renal insufficiency, complete urinary elimination may take much longer. However, due to the strong complex formation with rocuronium, no signs of recurrence of neuromuscular blockade have been noted up to 48 hours after use in such patients.

Uses of Neuromuscular Blocking Drugs A. Surgical Relaxation One of the most important applications of the neuromuscular blockers is in facilitating intracavitary surgery, especially in intra-abdominal and intrathoracic procedures. B. Endotracheal Intubation By relaxing the pharyngeal and laryngeal muscles, neuromuscular blocking drugs facilitate laryngoscopy and placement of an endotracheal tube. Endotracheal tube placement ensures an adequate airway and minimizes the risk of pulmonary aspiration during general anesthesia. C. Control of Ventilation In critically ill patients who have ventilatory failure from various causes (eg, severe bronchospasm, pneumonia, chronic obstructive airway disease), it may be necessary to control ventilation to provide adequate gas exchange and to prevent atelectasis. In the ICU, neuromuscular blocking drugs are frequently administered to reduce chest wall resistance (ie, improve thoracic compliance), decrease oxygen utilization, and improve ventilator synchrony. D. Treatment of Convulsions Neuromuscular blocking drugs (ie, succinylcholine) are occasionally used to attenuate the peripheral (motor) manifestations of convulsions associated with status epilepticus, local anesthetic toxicity, or electroconvulsive therapy. Although this approach is effective in eliminating the muscular manifestations of the seizures, it has no effect on the central processes because neuromuscular blocking drugs do not cross the blood-brain barrier.

SPASMOLYTIC DRUGS


Spasticity may be defined as “disordered sensorimotor control resulting from an upper motor neuron lesion, presenting as intermittent or sustained involuntary activation of muscles.” It is characterized by an increase in tonic stretch reflexes and flexor muscle spasms (ie, increased basal muscle tone) together with muscle weakness. It is often associated with spinal injury, cerebral palsy, multiple sclerosis, and stroke. These conditions often involve abnormal function of the bowel and bladder as well as skeletal muscle. As described by the definition above, the mechanisms underlying clinical spasticity appear to involve not only the stretch reflex arc itself but also higher centers in the CNS (ie, upper motor neuron lesion), with damage to descending pathways in the spinal cord resulting in hyperexcitability of the alpha motor neurons in the cord. Pharmacologic therapy may ameliorate some of the symptoms of spasticity by modifying the stretch reflex arc or by interfering directly with skeletal muscle (ie, excitation-contraction coupling). The important components involved in these processes are shown in Figure 27–9.

FIGURE 27–9 Schematic illustration of the structures involved in the stretch reflex (right half) showing innervation of extrafusal (striated muscle) fibers by alpha motor neurons and of intrafusal fibers (within muscle spindle) by gamma motor neurons. The left half of the diagram shows an inhibitory reflex arc, which includes an intercalated inhibitory interneuron. (Reproduced, with permission, from Waxman SG: Clinical Neuroanatomy, 26th edition. McGraw-Hill, 2009. Copyright © The McGraw-Hill Companies, Inc.) Drugs that modify the reflex arc may modulate excitatory or inhibitory synapses (see Chapter 21). Thus, to reduce the hyperactive stretch reflex, it is desirable to reduce the activity of the Ia fibers that excite the primary motor neuron or to enhance the activity of the inhibitory internuncial neurons. These structures are shown in greater detail in Figure 27–10.


FIGURE 27–10 Postulated sites of spasmolytic action of tizanidine (α2 ), benzodiazepines (GABAA), and baclofen (GABAB) in the spinal cord. Tizanidine may also have a postsynaptic inhibitory effect. Dantrolene acts on the sarcoplasmic reticulum in skeletal muscle. Glu, glutamatergic neuron. A variety of pharmacologic agents described as depressants of the spinal “polysynaptic” reflex arc (eg, barbiturates [phenobarbital] and glycerol ethers [mephenesin]) have been used to treat these conditions of excess skeletal muscle tone. However, as illustrated in Figure 27–10, nonspecific depression of synapses involved in the stretch reflex could reduce the desired GABAergic inhibitory activity, as well as the excitatory glutamatergic transmission. Currently available drugs can provide significant relief from painful muscle spasms, but they are less effective in improving meaningful function (eg, mobility and return to work).

Diazepam As described in Chapter 22, benzodiazepines facilitate the action of GABA in the CNS. Diazepam acts at GABA A synapses, and its


action in reducing spasticity is at least partly mediated in the spinal cord because it is somewhat effective in patients with cord transection. Although diazepam can be used in patients with muscle spasm of almost any origin (including local muscle trauma), it also produces sedation at the doses required to reduce muscle tone. The initial dosage is 4 mg/d, and it is gradually increased to a maximum of 60 mg/d. Other benzodiazepines have been used as spasmolytics (eg, midazolam), but clinical experience with them is limited.

Baclofen Baclofen (p-chlorophenyl-GABA) was designed to be an orally active GABA-mimetic agent and is an agonist at GABAB receptors. Activation of these receptors by baclofen results in hyperpolarization by three distinct actions: 1) closure of presynaptic calcium channels, 2) increased postsynaptic K+ conductance, and 3) inhibition of dendritic calcium influx channels (see Figure 24–2 and Figure 27–10). Through reduced release of excitatory transmitters in both the brain and the spinal cord, baclofen suppresses activity of Ia sensory afferents, spinal interneurons, and motor neurons. Baclofen may also reduce pain in patients with spasticity, perhaps by inhibiting the release of substance P (neurokinin-1) in the spinal cord.

Baclofen is at least as effective as diazepam in reducing spasticity and causes less sedation. In addition, baclofen does not reduce overall muscle strength as much as dantrolene. It is rapidly and completely absorbed after oral administration and has a plasma half-life of 3–4 hours. Dosage is started at 15 mg twice daily, increasing as tolerated to 100 mg daily. Adverse effects of this drug include drowsiness; however, patients become tolerant to the sedative effect with chronic administration. Increased seizure activity has been reported in epileptic patients. Therefore, withdrawal from baclofen must be done very slowly. Baclofen should be used with caution during pregnancy: although there are no reports of baclofen directly causing human fetal malformations, animal studies using high doses show that it causes impaired sternal ossification and omphalocele. Studies have confirmed that intrathecal administration of baclofen can control severe spasticity and muscle pain that is not responsive to medication by other routes of administration. Owing to the poor egress of baclofen from the spinal cord, peripheral symptoms are rare. Therefore, higher central concentrations of the drug may be tolerated. Partial tolerance to the effect of the drug may occur after several months of therapy, but can be overcome by upward dosage adjustments to maintain the beneficial effect. This tolerance was not confirmed in a recent study and decreased response may represent unrecognized catheter malfunctions. Although a major disadvantage of this therapeutic approach is the difficulty of maintaining the drug delivery catheter in the subarachnoid space, risking an acute withdrawal syndrome upon treatment interruption, long-term intrathecal baclofen therapy can improve the quality of life for patients with severe spastic disorders. Adverse effects of high-dose baclofen include excessive somnolence, respiratory depression, and coma. Oral baclofen has been studied in many other medical conditions, including patients with intractable low back pain, stiff person syndrome, trigeminal neuralgia, cluster headache, intractable hiccups, tic disorder, gastroesophageal reflux disease, and cravings for alcohol, nicotine, and cocaine (see Chapter 32).

TIZANIDINE As noted in Chapter 11, α2 agonists such as clonidine and other imidazoline compounds have a variety of effects on the CNS that are not fully understood. Among these effects is the ability to reduce muscle spasm. Tizanidine is a congener of clonidine that has been studied for its spasmolytic actions. Tizanidine has significant α 2 -adrenoceptor agonist effects, but it reduces spasticity in experimental models at doses that cause fewer cardiovascular effects than clonidine or dexmedetomidine. Tizanidine has approximately one tenth to one fifteenth of the blood pressure-lowering effects of clonidine. Neurophysiologic studies in animals and humans suggest that tizanidine reinforces both presynaptic and postsynaptic inhibition in the cord. It also inhibits nociceptive transmission in the spinal dorsal horn. Tizanidine’s actions are believed to be mediated via restoration of inhibitory suppression of the group II spinal interneurons without inducing any changes in intrinsic muscle properties. Clinical trials with oral tizanidine report efficacy in relieving muscle spasm comparable to diazepam, baclofen, and dantrolene. Tizanidine causes markedly less muscle weakness but produces a different spectrum of adverse effects, including drowsiness, hypotension, dizziness, dry mouth, asthenia, and hepatotoxicity. The drowsiness can be managed by taking the drug at night. Tizanidine displays linear pharmacokinetics, and dosage requirements vary considerably among patients. Dosage must be adjusted in patients with hepatic or renal impairment. Tizanidine is involved in drug-drug interactions; plasma levels increase in response to CYP1A2 inhibition. Conversely, tizanidine induces CYP11A1 activity, which is responsible for converting cholesterol to pregnenolone. In addition to its


effectiveness in spastic conditions, tizanidine also appears to be effective for management of chronic migraine.

OTHER CENTRALLY ACTING SPASMOLYTIC DRUGS Gabapentin is an antiepileptic drug (see Chapter 24) that has shown considerable promise as a spasmolytic agent in several studies involving patients with multiple sclerosis. Pregabalin is a newer analog of gabapentin that may also prove useful in relieving painful disorders that involve a muscle spasm component. Progabide and glycine have also been found in preliminary studies to reduce spasticity. Progabide is a GABA A and GABAB agonist and has active metabolites, including GABA itself. Glycine is another inhibitory amino acid neurotransmitter (see Chapter 21) that appears to possess pharmacologic activity when given orally and readily passes the blood-brain barrier. Idrocilamide and riluzole are newer drugs for the treatment of amyotrophic lateral sclerosis (ALS) that appear to have spasm-reducing effects, possibly through inhibition of glutamatergic transmission in the CNS.

DANTROLENE Dantrolene is a hydantoin derivative related to phenytoin that has a unique mechanism of spasmolytic activity. In contrast to the centrally acting drugs, dantrolene reduces skeletal muscle strength by interfering with excitation-contraction coupling in the muscle fibers. The normal contractile response involves release of calcium from its stores in the sarcoplasmic reticulum (see Figures 13–1 and 27–10). This activator calcium brings about the tension-generating interaction of actin with myosin. Calcium is released from the sarcoplasmic reticulum via a calcium channel, called the ryanodine receptor (RyR) channel because the plant alkaloid ryanodine combines with a receptor on the channel protein. In the case of the skeletal muscle RyR1 channel, ryanodine facilitates the open configuration.

Dantrolene interferes with the release of activator calcium through this sarcoplasmic reticulum calcium channel by binding to the RyR1 and blocking the opening of the channel. Motor units that contract rapidly are more sensitive to the drug’s effects than are slowerresponding units. Cardiac muscle and smooth muscle are minimally depressed because the release of calcium from their sarcoplasmic reticulum involves a different RyR channel (RyR2). Treatment with dantrolene is usually initiated with 25 mg daily as a single dose, increasing to a maximum of 100 mg four times daily as tolerated. Only about one third of an oral dose of dantrolene is absorbed, and the elimination half-life of the drug is approximately 8 hours. Major adverse effects are generalized muscle weakness, sedation, and occasionally hepatitis. A special application of dantrolene is in the treatment of malignant hyperthermia, a rare heritable disorder that can be triggered by a variety of stimuli, including general anesthetics (eg, volatile anesthetics) and neuromuscular blocking drugs (eg, succinylcholine; see also Chapter 16). Patients at risk for this condition have a hereditary alteration in Ca2+-induced Ca2+ release via the RyR1 channel or impairment in the ability of the sarcoplasmic reticulum to sequester calcium via the Ca2+ transporter (Figure 27–10). Several mutations associated with this risk have been identified. After administration of one of the triggering agents, there is a sudden and prolonged release of calcium, with massive muscle contraction, lactic acid production, and increased body temperature. Prompt treatment is essential to control acidosis and body temperature and to reduce calcium release. The latter is accomplished by administering intravenous dantrolene, starting with a dose of 1 mg/kg IV, and repeating as necessary to a maximum dose of 10 mg/kg.

BOTULINUM TOXIN The therapeutic use of botulinum toxin (BoNT) for ophthalmic purposes and for local muscle spasm was mentioned in Chapter 6. This neurotoxin produces chemodenervation and local paralysis when injected into a muscle. Seven immunologically distinct toxins share homologous subunits. The single-chain polypeptide BoNT has little activity until it is cleaved into a heavy chain (100 kDa) and a light chain (50 kDa). The light chain, a zinc-dependent protease, prevents release of acetylcholine by interfering with vesicle fusion, through proteolytically cleaving SNAP * -25 (BoNT-A, BoNT-E) or synaptobrevin-2 (BoNT-B, BoNT-D, BoNT-F). Local facial injections of botulinum toxin are widely used for the short-term treatment (1–3 months per treatment) of wrinkles associated with aging around the eyes and mouth. Local injection of botulinum toxin has also become a useful treatment for generalized spastic disorders (eg, cerebral palsy). Most clinical studies to date have involved administration in one or two limbs, and the benefits appear to persist for weeks to


several months after a single treatment. BoNT has virtually replaced anticholinergic medications used in the treatment of dystonia. More recently, FDA approval was granted for treatment of incontinence due to overactive bladder and for chronic migraine. Most studies have used several formulations of type A BoNT, but type B is also available. Adverse effects include respiratory tract infections, muscle weakness, urinary incontinence, falls, fever, and pain. While immunogenicity is currently of much less concern than in the past, experts still recommend that injections not be administered more frequently than every 3 months. Studies to determine safety of more frequent administration are underway. Besides occasional complications, a major limitation of BoNT treatment is its high cost. Future research developing other serotypes such as BoNT-C and BoNT-F is expected to result in the development of new agents that can provide chemodenervation with long-term benefits and at lower cost.

DRUGS USED TO TREAT ACUTE LOCAL MUSCLE SPASM A large number of less well-studied, centrally active drugs (eg, carisoprodol, chlorphenesin, chlorzoxazone, cyclobenzaprine, metaxalone, methocarbamol, and orphenadrine) are promoted for the relief of acute muscle spasm caused by local tissue trauma or muscle strains. It has been suggested that these drugs act primarily at the level of the brainstem. Cyclobenzaprine may be regarded as the prototype of the group. Cyclobenzaprine is structurally related to the tricyclic antidepressants and produces antimuscarinic side effects. It is ineffective in treating muscle spasm due to cerebral palsy or spinal cord injury. As a result of its strong antimuscarinic actions, cyclobenzaprine may cause significant sedation, as well as confusion and transient visual hallucinations. The dosage of cyclobenzaprine for acute injury-related muscle spasm is 20–40 mg/d orally in divided doses. *

The authors thank Paul F. White, PhD, MD, and Bertram G. Katzung, MD, PhD, for contributions to this chapter in previous editions.

*

SNAP, Soluble N-ethylmaleimide sensitive factor Attachment Protein.

SUMMARY Skeletal Muscle Relaxants



PREPARATIONS AVAILABLE

REFERENCES Neuromuscular Blockers Belmont MR et al: Clinical pharmacology of GW280430A in humans. Anesthesiology 2004;100:768. Brull SJ, Murphy GS: Residual neuromuscular block: Lessons unlearned. Part II: Methods to reduce the risk of residual weakness. Anesth Analg 2010;111:129. De Boer HD et al: Reversal of rocuronium-induced (1.2 mg/kg) profound neuromuscular blockade by sugammadex. Anesthesiology 2007;107:239. Gibb AJ, Marshall IG: Pre- and postjunctional effects of tubocurarine and other nicotinic antagonists during repetitive stimulation in the rat. J Physiol 1984;351:275. Hemmerling T M, Russo G, Bracco D: Neuromuscular blockade in cardiac surgery: An update for clinicians. Ann Card Anaesth 2008;11:80. Hirsch NP: Neuromuscular junction in health and disease. Br J Anaesth 2007;99:132. Kampe S et al: Muscle relaxants. Best Prac Res Clin Anesthesiol 2003;17:137. Lee C: Structure, conformation, and action of neuromuscular blocking drugs. Br J Anaesth 2001;87:755. Lee C et al: Reversal of profound neuromuscular block by sugammadex administered three minutes after rocuronium. Anesthesiology 2009;110:1020. Lien CA et al: Fumarates: Unique nondepolarizing neuromuscular blocking agents that are antagonized by cysteine. J Crit Care 2009;24:50. Llauradรณ S et al: Sugammadex ideal body weight dose adjusted by level of neuromuscular blockade in laparoscopic bariatric surgery. Anesthesiology 2012;117:93.


Mace SE: Challenges and advances in intubation: rapid sequence intubation. Emerg Med Clin North Am 2008;26:1043. Marshall CG, Ogden DC, Colquhoun D: T he actions of suxamethonium (succinyldicholine) as an agonist and channel blocker at the nicotinic receptor of frog muscle. J Physiol (Lond) 1990;428:155. Martyn JA: Neuromuscular physiology and pharmacology. In: Miller RD (editor): Anesthesia, 7th ed. Churchill Livingstone, 2010. Meakin GH: Recent advances in myorelaxant therapy. Paed Anaesthesia 2001;11:523. Murphy GS, Brull SJ: Residual neuromuscular block: Lessons unlearned. Part I: Definitions, incidence, and adverse physiologic effects of residual neuromuscular block. Anesth Analg 2010;111:120. Naguib M: Sugammadex: Another milestone in clinical neuromuscular pharmacology. Anesth Analg 2007;104:575. Naguib M, Brull SJ: Update on neuromuscular pharmacology. Curr Opin Anaesthesiol 2009;22:483. Naguib M, Kopman AF, Ensor JE: Neuromuscular monitoring and postoperative residual curarisation: A meta-analysis. Br J Anaesth 2007;98:302. Naguib M et al: Advances in neurobiology of the neuromuscular junction: Implications for the anesthesiologist. Anesthesiology 2002;96:202. Nicholson WT , Sprung J, Jankowski CJ: Sugammadex: A novel agent for the reversal of neuromuscular blockade. Pharmacotherapy 2007;27:1181. Pavlin JD, Kent CD: Recovery after ambulatory anesthesia. Curr Opin Anaesthesiol 2008;21:729. Puhringer FK et al: Reversal of profound, high-dose rocuronium-induced neuromuscular blockade by sugammadex at two different time points. Anesthesiology 2008;109:188. Sacan O, Klein K, White PF: Sugammadex reversal of rocuronium-induced neuromuscular blockade: A comparison with neostigmine-glycopyrrolate and edrophoniumatropine. Anesth Analg 2007;104:569. Savarese JJ et al: Preclinical pharmacology of GW280430A (AV430A) in the rhesus monkey and in the cat: A comparison with mivacurium. Anesthesiology 2004;100:835. Sine SM: End-plate acetylcholine receptor: Structure, mechanism, pharmacology, and disease. Physiol Rev 2012;92:1189. Staals LM et al: Reduced clearance of rocuronium and sugammadex in patients with severe to end-stage renal failure: A pharmacokinetic study. Br J Anaesth 2010;104:31. Sunaga H et al: Gantacurium and CW002 do not potentiate muscarinic receptor-mediated airway smooth muscle constriction in guinea pigs. Anesthesiology 2010;112:892. Viby-Mogensen J: Neuromuscular monitoring. In: Miller RD (editor): Anesthesia, 5th ed. Churchill Livingstone, 2000.

Spasmolytics Caron E, Morgan R, Wheless JW: An unusual cause of flaccid paralysis and coma: Baclofen overdose. J Child Neurol 2014;29:555. Corcia P, Meininger V: Management of amyotrophic lateral sclerosis. Drugs 2008;68:1037. Cutter NC et al: Gabapentin effect on spasticity in multiple sclerosis: A placebo-controlled, randomized trial. Arch Phys Med Rehabil 2000;81:164. Draulans N et al: Intrathecal baclofen in multiple sclerosis and spinal cord injury: Complications and long-term dosage evolution. Clin Rehabil 2013;27:1137. Gracies JM, Singer BJ, Dunne JW: T he role of botulinum toxin injections in the management of muscle overactivity of the lower limb. Disabil Rehabil 2007;29:1789. Groves L, Shellenberger MK, Davis CS: T izanidine treatment of spasticity: A meta-analysis of controlled, double-blind, comparative studies with baclofen and diazepam. Adv T her 1998;15:241. Jankovic J: Medical treatment of dystonia. Mov Disord 2013;28:1001. Kheder A, Nair KPS: Spasticity: Pathophysiology, evaluation and management. Pract Neurol 2012;12:289. Krause T et al: Dantrolene—A review of its pharmacology, therapeutic use and new developments. Anaesthesia 2004;59:364. Lopez JR et al: Effects of dantrolene on myoplasmic free [Ca2+] measured in vivo in patients susceptible to malignant hyperthermia. Anesthesiology 1992;76:711. Lovell BV, Marmura MJ: New therapeutic developments in chronic migraine. Curr Opin Neurol 2010;23:254. Malanga G, Reiter RD, Garay E: Update on tizanidine for muscle spasticity and emerging indications. Expert Opin Pharmacother 2008;9:2209. Mast N, Linger M, Pikuleva IA: Inhibition and stimulation of activity of purified recombinant CYP11A1 by therapeutic agents. Mol Cell Endocrinol 2013;371:100. Mirbagheri MM, Chen D, Rymer WZ: Quantification of the effects of an alpha-2 adrenergic agonist on reflex properties in spinal cord injury using a system identification technique. J Neuroeng Rehabil 2010;7:29. Neuvonen PJ: T owards safer and more predictable drug treatment—Reflections from studies of the First BCPT Prize awardee. Basic Clin Pharmacol T oxicol 2012;110:207. Nolan KW, Cole LL, Liptak GS: Use of botulinum toxin type A in children with cerebral palsy. Phys T her 2006;86:573. Ronan S, Gold JT : Nonoperative management of spasticity in children. Childs Nerv Syst 2007;23:943. Ross JC et al: Acute intrathecal baclofen withdrawal: A brief review of treatment options. Neurocrit Care 2011;14:103. Vakhapova V, Auriel E, Karni A: Nightly sublingual tizanidine HCl in multiple sclerosis: Clinical efficacy and safety. Clin Neuropharmacol 2010;33:151. Verrotti A et al: Pharmacotherapy of spasticity in children with cerebral palsy. Pediatr Neurol 2006;34:1. Ward AB: Spasticity treatment with botulinum toxins. J Neural T ransm 2008;115:607.

CASE STUDY ANSWER Because of trauma and associated pain, it is assumed that gastric emptying will be significantly delayed. To avoid possible aspiration at the time of intubation, a very rapid-acting muscle relaxant should be used so the airway can be secured with an endotracheal tube. Therefore, succinylcholine is the agent of choice in this case. Despite its adverse effects, succinylcholine has the fastest onset of action of any currently available skeletal muscle relaxant. An alternative to succinylcholine is high-dose (up to 1.2 mg/kg) rocuronium, a nondepolarizing muscle relaxant. At this dose, rocuronium has a very rapid onset, which approaches but does not quite equal that of succinylcholine. Both burns and neurologic injuries result in the expression of extrajunctional acetylcholine receptors. In patients with recent burns, succinylcholine use can lead to life-threatening hyperkalemia. Although the drug would not result in dangerous hyperkalemia if given immediately after a severe neurologic injury, in a patient with a chronic paralysis, its use may lead to hyperkalemia. Therefore, succinylcholine would also be contraindicated in a patient with long-standing hemiparesis.



CHAPTER

28 Pharmacologic Management of Parkinsonism & Other Movement Disorders Michael J. Aminoff, MD, DSc, FRCP

CASE STUDY A 64-year-old architect complains of left-hand tremor at rest, which interferes with his writing and drawing. He also notes a stooped posture, a tendency to drag his left leg when walking, and slight unsteadiness on turning. He remains independent in all activities of daily living. Examination reveals hypomimia (flat facies), hypophonia, a rest tremor of the left arm and leg, mild rigidity in all limbs, and impaired rapid alternating movements in the left limbs. Neurologic and general examinations are otherwise normal. What is the likely diagnosis and prognosis? He is started on a dopamine agonist, which he seems to tolerate well, and the dose is gradually built up to the therapeutic range. About a year later, he and his wife return for follow-up. It now becomes apparent that he is spending large sums of money, which he cannot afford, on gambling and refuses to stop, despite his wife’s entreaties. To what is his condition due and how should it be managed?

Several types of abnormal movement are recognized. Tremor consists of a rhythmic oscillatory movement around a joint and is best characterized by its relation to activity. Tremor at rest is characteristic of parkinsonism, when it is often associated with rigidity and an impairment of voluntary activity. Tremor may occur during maintenance of sustained posture (postural tremor) or during movement (intention tremor). A conspicuous postural tremor is the cardinal feature of benign essential or familial tremor. Intention tremor occurs in patients with a lesion of the brainstem or cerebellum, especially when the superior cerebellar peduncle is involved; it may also occur as a manifestation of toxicity from alcohol or certain other drugs. Chorea consists of irregular, unpredictable, involuntary muscle jerks that occur in different parts of the body and impair voluntary activity. In some instances, the proximal muscles of the limbs are most severely affected, and because the abnormal movements are then particularly violent, the term ballismus has been used to describe them. Chorea may be hereditary or may occur as a complication of a number of general medical disorders and of therapy with certain drugs. Abnormal movements may be slow and writhing in character (athetosis) and in some instances are so sustained that they are more properly regarded as abnormal postures (dystonia). Athetosis or dystonia may occur with perinatal brain damage, with focal or generalized cerebral lesions, as an acute complication of certain drugs, as an accompaniment of diverse neurologic disorders, or as an isolated inherited phenomenon of uncertain cause known as idiopathic torsion dystonia or dystonia musculorum deformans. Various genetic loci have been reported depending on the age of onset, mode of inheritance, and response to dopaminergic therapy. The physiologic basis is uncertain, and treatment is unsatisfactory. Tics are sudden coordinated abnormal movements that tend to occur repetitively, particularly about the face and head, especially in children, and can be suppressed voluntarily for short periods of time. Common tics include repetitive sniffing or shoulder shrugging. Tics may be single or multiple and transient or chronic. Gilles de la Tourette’s syndrome is characterized by chronic multiple tics; its pharmacologic management is discussed at the end of this chapter. Many of the movement disorders have been attributed to disturbances of the basal ganglia. The basic circuitry of the basal ganglia involves three interacting neuronal loops that include the cortex and thalamus as well as the basal ganglia themselves (Figure 28–1). However, the precise function of these anatomic structures is not yet fully understood, and it is not possible to relate individual symptoms to involvement at specific sites.


FIGURE 28–1 Functional circuitry between the cortex, basal ganglia, and thalamus. The major neurotransmitters are indicated. In Parkinson’s disease, there is degeneration of the pars compacta of the substantia nigra, leading to overactivity in the indirect pathway (red) and increased glutamatergic activity by the subthalamic nucleus.

PARKINSONISM Parkinsonism is characterized by a combination of rigidity, bradykinesia, tremor, and postural instability that can occur for a variety of reasons but is usually idiopathic (Parkinson’s disease or paralysis agitans). Cognitive decline occurs in many patients as the disease advances. Other non-motor symptoms—which are receiving increasing attention—are affective disorders (anxiety or depression), personality changes, abnormalities of autonomic function (sphincter or sexual functions; choking; sweating abnormalities; and disturbances of blood pressure regulation), sleep disorders, and sensory complaints or pain. The disease is generally progressive, leading to increasing disability unless effective treatment is provided.

Pathogenesis The pathogenesis of parkinsonism seems to relate to a combination of impaired degradation of proteins, intracellular protein accumulation and aggregation, oxidative stress, mitochondrial damage, inflammatory cascades, and apoptosis. Studies in twins suggest that genetic


factors are important, especially when the disease occurs in patients under age 50. Recognized genetic abnormalities account for 10– 15% of cases. Mutations of the α-synuclein gene at 4q21 or duplication and triplication of the normal synuclein gene are associated with Parkinson’s disease, which is now widely recognized as a synucleinopathy. Mutations of the leucine-rich repeat kinase 2 (LRRK2) gene at 12cen, and the UCHL1 gene may also cause autosomal dominant parkinsonism. Mutations in the parkin gene (6q25.2–q27) cause early onset, autosomal recessive, familial parkinsonism, or sporadic juvenile-onset parkinsonism. Several other genes or chromosomal regions have been associated with familial forms of the disease. Environmental or endogenous toxins may also be important in the etiology of the disease. Epidemiologic studies reveal that cigarette smoking, coffee, anti-inflammatory drug use, and high serum uric acid levels are protective, whereas the incidence of the disease is increased in those working in teaching, health care, or farming, and in those with lead or manganese exposure or with vitamin D deficiency. The finding of Lewy bodies (intracellular inclusion bodies containing α-synuclein) in fetal dopaminergic cells transplanted into the brain of parkinsonian patients some years previously has provided some support for suggestions that Parkinson’s disease may represent a prion disease. Staining for α-synuclein has revealed that pathology is more widespread than previously recognized, developing initially in the olfactory nucleus and lower brainstem (stage 1 of Braak scale), then the higher brainstem (stage 2), the substantia nigra (stage 3), the mesocortex and thalamus (stage 4), and finally the entire neocortex (stage 5). The motor features of Parkinson’s disease develop at stage 3 on the Braak scale. The normally high concentration of dopamine in the basal ganglia of the brain is reduced in parkinsonism, and pharmacologic attempts to restore dopaminergic activity with levodopa and dopamine agonists alleviate many of the motor features of the disorder. An alternative but complementary approach has been to restore the normal balance of cholinergic and dopaminergic influences on the basal ganglia with antimuscarinic drugs. The pathophysiologic basis for these therapies is that in idiopathic parkinsonism, there is a loss of dopaminergic neurons in the substantia nigra that normally inhibit the output of GABAergic cells in the corpus striatum (Figure 28–2). Drugs that induce parkinsonian syndromes either are dopamine receptor antagonists (eg, antipsychotic agents; see Chapter 29) or lead to the destruction of the dopaminergic nigrostriatal neurons (eg, 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine [MPTP]; see below). Various other neurotransmitters, such as norepinephrine, are also depleted in the brain in parkinsonism, but these deficiencies are of uncertain clinical relevance.

FIGURE 28–2 Schematic representation of the sequence of neurons involved in parkinsonism. Top: Dopaminergic neurons (red) originating in the substantia nigra normally inhibit the GABAergic output from the striatum, whereas cholinergic neurons (green) exert an excitatory effect. Bottom: In parkinsonism, there is a selective loss of dopaminergic neurons (dashed, red).

LEVODOPA Dopamine does not cross the blood-brain barrier and if given into the peripheral circulation has no therapeutic effect in parkinsonism.


However, (−)-3-(3,4-dihydroxyphenyl)-L-alanine (levodopa), the immediate metabolic precursor of dopamine, does enter the brain (via an L-amino acid transporter, LAT), where it is decarboxylated to dopamine (see Figure 6–5). Several noncatecholamine dopamine receptor agonists have also been developed and may lead to clinical benefit, as discussed in the text that follows. Dopamine receptors are discussed in detail in Chapters 21 and 29. They exist in five subtypes. D1 and D5 receptors are classified as the D1 receptor family based on genetic and biochemical factors; D2 , D3 , and D4 are grouped as belonging to the D2 receptor family. Dopamine receptors of the D1 type are located in the pars compacta of the substantia nigra and presynaptically on striatal axons coming from cortical neurons and from dopaminergic cells in the substantia nigra. The D2 receptors are located postsynaptically on striatal neurons and presynaptically on axons in the substantia nigra belonging to neurons in the basal ganglia. The benefits of dopaminergic antiparkinsonism drugs appear to depend mostly on stimulation of the D2 receptors. However, D 1- receptor stimulation may also be required for maximal benefit and one of the newer drugs is D3 selective. Dopamine agonist or partial agonist ergot derivatives such as lergotrile and bromocriptine that are powerful stimulators of the D2 receptors have antiparkinsonism properties, whereas certain dopamine blockers that are selective D2 antagonists can induce parkinsonism.

Chemistry Dopa is the amino acid precursor of dopamine and norepinephrine (discussed in Chapter 6). Its structure is shown in Figure 28–3. Levodopa is the levorotatory stereoisomer of dopa.


FIGURE 28–3 Some drugs used in the treatment of parkinsonism.

Pharmacokinetics Levodopa is rapidly absorbed from the small intestine, but its absorption depends on the rate of gastric emptying and the pH of the gastric contents. Ingestion of food delays the appearance of levodopa in the plasma. Moreover, certain amino acids from ingested food can compete with the drug for absorption from the gut and for transport from the blood to the brain. Plasma concentrations usually peak between 1 and 2 hours after an oral dose, and the plasma half-life is usually between 1 and 3 hours, although it varies considerably among individuals. About two thirds of the dose appears in the urine as metabolites within 8 hours of an oral dose, the main metabolic


products being 3-methoxy-4-hydroxyphenyl acetic acid (homovanillic acid, HVA) and dihydroxyphenylacetic acid (DOPAC). Unfortunately, only about 1–3% of administered levodopa actually enters the brain unaltered; the remainder is metabolized extracerebrally, predominantly by decarboxylation to dopamine, which does not penetrate the blood-brain barrier. Accordingly, levodopa must be given in large amounts when used alone. However, when given in combination with a dopa decarboxylase inhibitor that does not penetrate the blood-brain barrier, the peripheral metabolism of levodopa is reduced, plasma levels of levodopa are higher, plasma half-life is longer, and more dopa is available for entry into the brain (Figure 28–4). Indeed, concomitant administration of a peripheral dopa decarboxylase inhibitor such as carbidopa may reduce the daily requirements of levodopa by approximately 75%.


FIGURE 28–4 Fate of orally administered levodopa and the effect of carbidopa, estimated from animal data. The width of each pathway indicates the absolute amount of the drug at each site, whereas the percentages shown denote the relative proportion of the administered dose. The benefits of co-administration of carbidopa include reduction of the amount of levodopa required for benefit and of the absolute amount diverted to peripheral tissues and an increase in the fraction of the dose that reaches the brain. GI, gastrointestinal. (Data from Nutt JG, Fellman JH: Pharmacokinetics of levodopa. Clin Neuropharmacol 1984;7:35.)


Clinical Use The best results of levodopa treatment are obtained in the first few years of treatment. This is sometimes because the daily dose of levodopa must be reduced over time to avoid adverse effects at doses that were well tolerated initially. Some patients become less responsive to levodopa, perhaps because of loss of dopaminergic nigrostriatal nerve terminals or some pathologic process directly involving striatal dopamine receptors. For such reasons, the benefits of levodopa treatment often begin to diminish after about 3 or 4 years of therapy, regardless of the initial therapeutic response. Although levodopa therapy does not stop the progression of parkinsonism, its early initiation lowers the mortality rate. However, long-term therapy may lead to a number of problems in management such as the on-off phenomenon discussed below. The most appropriate time to introduce levodopa therapy must therefore be determined individually. When levodopa is used, it is generally given in combination with carbidopa (Figure 28–3), a peripheral dopa decarboxylase inhibitor, which reduces peripheral conversion to dopamine. Combination treatment is started with a small dose, eg, carbidopa 25 mg, levodopa 100 mg three times daily, and gradually increased. It should be taken 30–60 minutes before meals. Most patients ultimately require carbidopa 25 mg, levodopa 250 mg three or four times daily. It is generally preferable to keep treatment with this agent at a low level (eg, carbidopa-levodopa 25/100 three times daily) when possible, and if necessary, to add a dopamine agonist, to reduce the risk of development of response fluctuations. A controlled-release formulation of carbidopa-levodopa is available and may be helpful in patients with established response fluctuations or as a means of reducing dosing frequency. A formulation of carbidopa-levodopa (10/100, 25/100, 25/250) that disintegrates in the mouth and is swallowed with the saliva (Parcopa) is available commercially and is best taken about 1 hour before meals. The combination (Stalevo) of levodopa, carbidopa, and a catechol-O-methyltransferase (COMT) inhibitor (entacapone) is discussed in a later section. Finally, therapy by infusion of levodopa-carbidopa into the duodenum or upper jejunum appears to be safe and is superior to a number of oral combination therapies in patients with response fluctuations. This is an approved therapy in Europe and Canada for treating advanced levodopa-responsive parkinsonism but is not yet available in the USA. A permanent access tube is inserted via a percutaneous endoscopic gastrostomy in patients who have responded well to carbidopa-levodopa gel administered through a nasoduodenal tube. A morning bolus (100–300 mg of levodopa) is delivered via a portable infusion pump, followed by a continuous maintenance dose (40–120 mg/h), with supplemental bolus doses as required. Levodopa can ameliorate many of the clinical motor features of parkinsonism, but it is particularly effective in relieving bradykinesia and any disabilities resulting from it. When it is first introduced, about one third of patients respond very well and one third less well. Most of the remainder either are unable to tolerate the medication or simply do not respond at all, especially if they do not have classic Parkinson’s disease.

Adverse Effects A. Gastrointestinal Effects When levodopa is given without a peripheral decarboxylase inhibitor, anorexia and nausea and vomiting occur in about 80% of patients. These adverse effects can be minimized by taking the drug in divided doses, with or immediately after meals, and by increasing the total daily dose very slowly. Antacids taken 30–60 minutes before levodopa may also be beneficial. The vomiting has been attributed to stimulation of the chemoreceptor trigger zone located in the brainstem but outside the blood-brain barrier. Fortunately, tolerance to this emetic effect develops in many patients. Antiemetics such as phenothiazines should be avoided because they reduce the antiparkinsonism effects of levodopa and may exacerbate the disease. When levodopa is given in combination with carbidopa, adverse gastrointestinal effects are much less frequent and troublesome, occurring in less than 20% of cases, so that patients can tolerate proportionately higher doses. B. Cardiovascular Effects A variety of cardiac arrhythmias have been described in patients receiving levodopa, including tachycardia, ventricular extrasystoles and, rarely, atrial fibrillation. This effect has been attributed to increased catecholamine formation peripherally. The incidence of such arrhythmias is low, even in the presence of established cardiac disease, and may be reduced still further if the levodopa is taken in combination with a peripheral decarboxylase inhibitor. Postural hypotension is common, but often asymptomatic, and tends to diminish with continuing treatment. Hypertension may also occur, especially in the presence of nonselective monoamine oxidase inhibitors or sympathomimetics or when massive doses of levodopa are being taken. C. Behavioral Effects A wide variety of adverse mental effects have been reported, including depression, anxiety, agitation, insomnia, somnolence, confusion, delusions, hallucinations, nightmares, euphoria, and other changes in mood or personality. Such adverse effects are more common in patients taking levodopa in combination with a decarboxylase inhibitor rather than levodopa alone, presumably because higher levels are reached in the brain. They may be precipitated by intercurrent illness or operation. It may be necessary to reduce or withdraw the medication. Several atypical antipsychotic agents that have low affinity for dopamine D2 receptors (clozapine, olanzapine, quetiapine, and risperidone; see Chapter 29) are now available and may be particularly helpful in counteracting such behavioral complications.


D. Dyskinesias and Response Fluctuations Dyskinesias occur in up to 80% of patients receiving levodopa therapy for more than 10 years. The character of dopa dyskinesias varies between patients but tends to remain constant in individual patients. Choreoathetosis of the face and distal extremities is the most common presentation. The development of dyskinesias is dose related, but there is considerable individual variation in the dose required to produce them. A number of compounds are being studied as possible antidyskinetic agents, but these studies are still at an early stage. Certain fluctuations in clinical response to levodopa occur with increasing frequency as treatment continues. In some patients, these fluctuations relate to the timing of levodopa intake (wearing-off reactions or end-of-dose akinesia). In other instances, fluctuations in clinical state are unrelated to the timing of doses (on-off phenomenon). In the on-off phenomenon, off-periods of marked akinesia alternate over the course of a few hours with on-periods of improved mobility but often marked dyskinesia. For patients with severe offperiods who are unresponsive to other measures, subcutaneously injected apomorphine may provide temporary benefit. The phenomenon is most likely to occur in patients who responded well to treatment initially. The exact mechanism is unknown. The dyskinesias may relate to an unequal distribution of striatal dopamine. Dopaminergic denervation plus chronic pulsatile stimulation of dopamine receptors with levodopa has been associated with development of dyskinesias. A lower incidence of dyskinesias occurs when levodopa is administered continuously (eg, intraduodenally or intrajejunally), and with drug delivery systems that enable a more continuous delivery of dopaminergic medication. E. Miscellaneous Adverse Effects Mydriasis may occur and may precipitate an attack of acute glaucoma in some patients. Other reported but rare adverse effects include various blood dyscrasias; a positive Coombs’ test with evidence of hemolysis; hot flushes; aggravation or precipitation of gout; abnormalities of smell or taste; brownish discoloration of saliva, urine, or vaginal secretions; priapism; and mild—usually transient— elevations of blood urea nitrogen and of serum transaminases, alkaline phosphatase, and bilirubin.

Drug Holidays A drug holiday (discontinuance of the drug for 3–21 days) may temporarily improve responsiveness to levodopa and alleviate some of its adverse effects but is usually of little help in the management of the on-off phenomenon. Furthermore, a drug holiday carries the risks of aspiration pneumonia, venous thrombosis, pulmonary embolism, and depression resulting from the immobility accompanying severe parkinsonism. For these reasons and because of the temporary nature of any benefit, drug holidays are not recommended.

Drug Interactions Pharmacologic doses of pyridoxine (vitamin B6 ) enhance the extracerebral metabolism of levodopa and may therefore prevent its therapeutic effect unless a peripheral decarboxylase inhibitor is also taken. Levodopa should not be given to patients taking monoamine oxidase A inhibitors or within 2 weeks of their discontinuance because such a combination can lead to hypertensive crises.

Contraindications Levodopa should not be given to psychotic patients because it may exacerbate the mental disturbance. It is also contraindicated in patients with angle-closure glaucoma, but those with chronic open-angle glaucoma may be given levodopa if intraocular pressure is well controlled and can be monitored. It is best given combined with carbidopa to patients with cardiac disease; even so, the risk of cardiac dysrhythmia is slight. Patients with active peptic ulcer must also be managed carefully, since gastrointestinal bleeding has occasionally occurred with levodopa. Because levodopa is a precursor of skin melanin and conceivably may activate malignant melanoma, it should be used with particular care in patients with a history of melanoma or with suspicious undiagnosed skin lesions; such patients should be monitored by a dermatologist regularly.

DOPAMINE RECEPTOR AGONISTS Drugs acting directly on postsynaptic dopamine receptors may have a beneficial effect in addition to that of levodopa (Figure 28–5). Unlike levodopa, they do not require enzymatic conversion to an active metabolite, act directly on the postsynaptic dopamine receptors, have no potentially toxic metabolites, and do not compete with other substances for active transport into the blood and across the bloodbrain barrier. Moreover, drugs selectively affecting certain (but not all) dopamine receptors may have more limited adverse effects than levodopa. A number of dopamine agonists have antiparkinsonism activity. The older dopamine agonists (bromocriptine and pergolide) are ergot (ergoline) derivatives (see Chapter 16), and are rarely—if ever—used to treat parkinsonism. Their side effects are of more concern than those of the newer agents (pramipexole and ropinirole). However, various impulse control disorders (such as gambling disorders, compulsive shopping, or hypersexuality) may be enhanced by activation of D2 or D3 dopamine receptors in the


mesocorticolimbic system in certain individuals. These may occur with one dopamine agonist and not another. They are not dosedependent, but in some patients a dose reduction may ameliorate them. The prevalence of impulse control disorders varies in different reports but may be as high as 15–25% in parkinsonian patients treated with these agents. Risk factors include a history of drug use or a family history of gambling disorders. There is no evidence that one agonist is superior to another; individual patients, however, may respond to one but not another of these agents. Moreover, their duration of action varies and is lengthened by extended-release preparations. Apomorphine is a potent dopamine agonist but is discussed separately in a later section in this chapter because it is used primarily as a rescue drug for patients with disabling response fluctuations to levodopa. Dopamine agonists have an important role as first-line therapy for Parkinson’s disease, and their use is associated with a lower incidence of the response fluctuations and dyskinesias that occur with long-term levodopa therapy. In consequence, dopaminergic therapy is often initiated with a dopamine agonist. Alternatively, a low dose of carbidopa plus levodopa (eg, Sinemet-25/100 three times daily) is introduced, and a dopamine agonist is then added. In either case, the dose of the dopamine agonist is built up gradually depending on response and tolerance. Dopamine agonists may also be given to patients with parkinsonism who are taking levodopa and who have end-of-dose akinesia or on-off phenomenon or are becoming resistant to treatment with levodopa. In such circumstances, it is generally necessary to lower the dose of levodopa to prevent intolerable adverse effects. The response to a dopamine agonist is generally disappointing in patients who have never responded to levodopa.

Bromocriptine Bromocriptine is a D2 agonist; its structure is shown in Table 16–6. This drug has been widely used to treat Parkinson’s disease in the past but is now rarely used for this purpose, having been superseded by the newer dopamine agonists. The usual daily dose of bromocriptine for parkinsonism varies between 7.5 and 30 mg. To minimize adverse effects, the dose is built up slowly over 2 or 3 months depending on response or the development of adverse reactions.

Pergolide Pergolide, another ergot derivative, directly stimulates both D1 and D2 receptors. It too has been widely used for parkinsonism but is no longer available in the United States because its use has been associated with the development of valvular heart disease. It is nevertheless still used in certain countries.

Pramipexole Pramipexole is not an ergot derivative, but it has preferential affinity for the D3 family of receptors. It is effective as monotherapy for mild parkinsonism and is also helpful in patients with advanced disease, permitting the dose of levodopa to be reduced and smoothing out response fluctuations. Pramipexole may ameliorate affective symptoms. A possible neuroprotective effect has been suggested by its ability to scavenge hydrogen peroxide and enhance neurotrophic activity in mesencephalic dopaminergic cell cultures.

Pramipexole is rapidly absorbed after oral administration, reaching peak plasma concentrations in approximately 2 hours, and is excreted largely unchanged in the urine. It is started at a dosage of 0.125 mg three times daily, doubled after 1 week, and again after another week. Further increments in the daily dose are by 0.75 mg at weekly intervals, depending on response and tolerance. Most patients require between 0.5 and 1.5 mg three times daily. Renal insufficiency may necessitate dosage adjustment. An extended-release preparation is now available and is taken once daily at a dose equivalent to the total daily dose of standard pramipexole. The extendedrelease preparation is generally more convenient for patients and avoids swings in blood levels of the drug over the day.

Ropinirole Another nonergoline derivative, ropinirole (now available in a generic preparation) is a relatively pure D2 receptor agonist that is effective as monotherapy in patients with mild disease and as a means of smoothing the response to levodopa in patients with more advanced disease and response fluctuations. It is introduced at 0.25 mg three times daily, and the total daily dose is then increased by 0.75 mg at


weekly intervals until the fourth week and by 1.5 mg thereafter. In most instances, a dosage between 2 and 8 mg three times daily is necessary. Ropinirole is metabolized by CYP1A2; other drugs metabolized by this isoform may significantly reduce its clearance. A prolonged-release preparation taken once daily is available.

Rotigotine The dopamine agonist rotigotine, delivered daily through a skin patch, is approved for treatment of early Parkinson’s disease. It supposedly provides more continuous dopaminergic stimulation than oral medication in early disease; its efficacy in more advanced disease is less clear. Benefits and side effects are similar to those of other dopamine agonists but reactions may also occur at the application site and are sometimes serious.

Adverse Effects of Dopamine Agonists A. Gastrointestinal Effects Anorexia and nausea and vomiting may occur when a dopamine agonist is introduced and can be minimized by taking the medication with meals. Constipation, dyspepsia, and symptoms of reflux esophagitis may also occur. Bleeding from peptic ulceration has been reported. B. Cardiovascular Effects Postural hypotension may occur, particularly at the initiation of therapy. Painless digital vasospasm is a dose-related complication of longterm treatment with the ergot derivatives (bromocriptine or pergolide). When cardiac arrhythmias occur, they are an indication for discontinuing treatment. Peripheral edema is sometimes problematic. Cardiac valvulopathy may occur with pergolide. C. Dyskinesias Abnormal movements similar to those introduced by levodopa may occur and are reversed by reducing the total dose of dopaminergic drugs being taken. D. Mental Disturbances Confusion, hallucinations, delusions, and other psychiatric reactions are potential complications of dopaminergic treatment and are more common and severe with dopamine receptor agonists than with levodopa. Disorders of impulse control may occur either as an exaggeration of a previous tendency or as a new phenomenon and may lead to compulsive gambling, shopping, betting, sexual activity, and other behaviors (see Chapter 32). They clear on withdrawal of the offending medication and sometimes simply with dose reductions. There appears to be no difference between the various dopamine agonists in their ability to induce these disorders. Impulse control disorders are generally under-reported by patients and their families and often unrecognized by health care professionals. E. Miscellaneous Headache, nasal congestion, increased arousal, pulmonary infiltrates, pleural and retroperitoneal fibrosis, and erythromelalgia are other reported adverse effects of the ergot-derived dopamine agonists. Erythromelalgia consists of red, tender, painful, swollen feet and, occasionally, hands, at times associated with arthralgia; symptoms and signs clear within a few days of withdrawal of the causal drug. In rare instances, an uncontrollable tendency to fall asleep at inappropriate times has occurred, particularly in patients receiving pramipexole or ropinirole; this requires discontinuation of the medication.

Contraindications Dopamine agonists are contraindicated in patients with a history of psychotic illness or recent myocardial infarction, or with active peptic ulceration. The ergot-derived agonists are best avoided in patients with peripheral vascular disease.


MONOAMINE OXIDASE INHIBITORS Two types of monoamine oxidase have been distinguished in the nervous system. Monoamine oxidase A metabolizes norepinephrine, serotonin, and dopamine; monoamine oxidase B metabolizes dopamine selectively. Selegiline (deprenyl) (Figure 28–3), a selective irreversible inhibitor of monoamine oxidase B at normal doses (at higher doses it inhibits monoamine oxidase A as well), retards the breakdown of dopamine (Figure 28–5); in consequence, it enhances and prolongs the antiparkinsonism effect of levodopa (thereby allowing the dose of levodopa to be reduced) and may reduce mild on-off or wearing-off phenomena. It is therefore used as adjunctive therapy for patients with a declining or fluctuating response to levodopa. The standard dose of selegiline is 5 mg with breakfast and 5 mg with lunch. Selegiline may cause insomnia when taken later during the day.

FIGURE 28–5 Pharmacologic strategies for dopaminergic therapy of Parkinson’s disease. Drugs and their effects are indicated (see text). MAO, monoamine oxidase; COMT, catechol-O-methyltransferase; DOPAC, dihydroxyphenylacetic acid; L-DOPA, levodopa; 3OMD, 3-O-methyldopa; 3-MT, 3-methoxytyramine. Selegiline has only a minor therapeutic effect on parkinsonism when given alone. Studies in animals suggest that it may reduce disease progression, but trials to test the effect of selegiline on the progression of parkinsonism in humans have yielded ambiguous results. The findings in a large multicenter study were taken to suggest a beneficial effect in slowing disease progression but may simply have reflected a symptomatic response. Rasagiline, another monoamine oxidase B inhibitor, is more potent than selegiline in preventing MPTP-induced parkinsonism and is


being used for early symptomatic treatment. The standard dosage is 1 mg/d. Rasagiline is also used as adjunctive therapy at a dosage of 0.5 or 1 mg/d to prolong the effects of levodopa-carbidopa in patients with advanced disease. A large double-blind, placebo-controlled, delayed-start study (the ADAGIO trial) to evaluate whether it had neuroprotective benefit (ie, slowed the disease course) yielded unclear results: a daily dose of 1 mg met all the end points of the study and did seem to slow disease progression, but a 2-mg dose failed to do so. These findings are difficult to explain and the decision to use rasagiline for neuroprotective purposes therefore remains an individual one. Neither selegiline nor rasagiline should be taken by patients receiving meperidine, tramadol, methadone, propoxyphene, cyclobenzaprine, or St. John’s wort. The antitussive dextromethorphan should also be avoided by patients taking one of the monoamine oxidase B inhibitors; indeed, it is wise to advise patients to avoid all over-the-counter cold preparations. Rasagiline or selegiline should be used with care in patients receiving tricyclic antidepressants or serotonin reuptake inhibitors because of the theoretical risk of acute toxic interactions of the serotonin syndrome type (see Chapter 16), but this is rarely encountered in practice. The adverse effects of levodopa may be increased by these drugs. The combined administration of levodopa and an inhibitor of both forms of monoamine oxidase (ie, a nonselective inhibitor) must be avoided, because it may lead to hypertensive crises, probably because of the peripheral accumulation of norepinephrine.

CATECHOL-O-METHYLTRANSFERASE INHIBITORS Inhibition of dopa decarboxylase is associated with compensatory activation of other pathways of levodopa metabolism, especially catechol-O-methyltransferase (COMT), and this increases plasma levels of 3-O-methyldopa (3-OMD). Elevated levels of 3-OMD have been associated with a poor therapeutic response to levodopa, perhaps in part because 3-OMD competes with levodopa for an active carrier mechanism that governs its transport across the intestinal mucosa and the blood-brain barrier. Selective COMT inhibitors such as tolcapone and entacapone also prolong the action of levodopa by diminishing its peripheral metabolism (Figure 28–5). Levodopa clearance is decreased, and relative bioavailability of levodopa is thus increased. Neither the time to reach peak concentration nor the maximal concentration of levodopa is increased. These agents may be helpful in patients receiving levodopa who have developed response fluctuations—leading to a smoother response, more prolonged on-time, and the option of reducing total daily levodopa dose. Tolcapone and entacapone are both widely available, but entacapone is generally preferred because it has not been associated with hepatotoxicity. The pharmacologic effects of tolcapone and entacapone are similar, and both are rapidly absorbed, bound to plasma proteins, and metabolized before excretion. However, tolcapone has both central and peripheral effects, whereas the effect of entacapone is peripheral. The half-life of both drugs is approximately 2 hours, but tolcapone is slightly more potent and has a longer duration of action. Tolcapone is taken in a standard dosage of 100 mg three times daily; some patients require a daily dose of twice that amount. By contrast, entacapone (200 mg) needs to be taken with each dose of levodopa, up to six times daily. Adverse effects of the COMT inhibitors relate in part to increased levodopa exposure and include dyskinesias, nausea, and confusion. It is often necessary to lower the daily dose of levodopa by about 30% in the first 48 hours to avoid or reverse such complications. Other adverse effects include diarrhea, abdominal pain, orthostatic hypotension, sleep disturbances, and an orange discoloration of the urine. Tolcapone may cause an increase in liver enzyme levels and has been associated rarely with death from acute hepatic failure; accordingly, it should not be used in patients with abnormal liver function test results. Its use in the USA requires signed patient consent (as provided in the product labeling) plus monitoring of liver function tests every 2–4 weeks during the first 6 months and periodically but less frequently thereafter. The medication should be withdrawn and not reintroduced if hepatic damage becomes evident. No such toxicity has been reported with entacapone. The commercial preparation named Stalevo consists of a combination of levodopa with both carbidopa and entacapone. It is available in three strengths: Stalevo 50 (50 mg levodopa plus 12.5 mg carbidopa and 200 mg entacapone), Stalevo 100 (100 mg, 25 mg, and 200 mg, respectively), and Stalevo 150 (150 mg, 37.5 mg, and 200 mg). Use of this preparation simplifies the drug regimen and requires the consumption of fewer tablets than otherwise. Stalevo is priced at or below the price of its individual components. The combination agent may provide greater symptomatic benefit than levodopa-carbidopa alone. However, despite the convenience of a single combination preparation, use of Stalevo rather than levodopa-carbidopa has been associated with earlier occurrence and increased frequency of dyskinesia. An investigation as to whether the use of Stalevo is associated with an increased risk for cardiovascular events (myocardial infarction, stroke, cardiovascular death) is ongoing.

APOMORPHINE Subcutaneous injection of apomorphine hydrochloride (Apokyn), a potent nonergoline dopamine agonist that interacts with postsynaptic D2 receptors in the caudate nucleus and putamen, is effective for the temporary relief (“rescue”) of off-periods of akinesia in patients on optimized dopaminergic therapy. It is rapidly taken up in the blood and then the brain, leading to clinical benefit that begins within about 10 minutes of injection and persists for up to 2 hours. The optimal dose is identified by administering increasing test doses until adequate benefit is achieved or a maximum of 0.6 mL (6 mg) is reached, with the supine and standing blood pressures monitored before injection and then every 20 minutes for an hour after it. Most patients require a dose of 3–6 mL (3–6 mg), and this should be given usually no


more than about three times daily, but occasionally up to five times daily. Nausea is often troublesome, especially at the initiation of apomorphine treatment; accordingly, pretreatment with the antiemetic trimethobenzamide (300 mg three times daily) for 3 days is recommended before apomorphine is introduced and is then continued for at least 1 month, if not indefinitely. Other adverse effects include dyskinesias, drowsiness, insomnia, chest pain, sweating, hypotension, syncope, constipation, diarrhea, mental or behavioral disturbances, panniculitis, and bruising at the injection site. Apomorphine should be prescribed only by physicians familiar with its potential complications and interactions. It should not be used in patients taking serotonin 5HT3 antagonists because severe hypotension may result.

AMANTADINE Amantadine, an antiviral agent, was by chance found to have relatively weak antiparkinsonism properties. Its mode of action in parkinsonism is unclear, but it may potentiate dopaminergic function by influencing the synthesis, release, or reuptake of dopamine. It has been reported to antagonize the effects of adenosine at adenosine A 2A receptors, which may inhibit D2 receptor function. Release of catecholamines from peripheral stores has also been documented. Amantadine is an antagonist of the NMDA-type glutamate receptor, suggesting an antidyskinetic effect.

Pharmacokinetics Peak plasma concentrations of amantadine are reached 1–4 hours after an oral dose. The plasma half-life is between 2 and 4 hours, most of the drug being excreted unchanged in the urine.

Clinical Use Amantadine is less efficacious than levodopa, and its benefits may be short-lived, often disappearing after only a few weeks of treatment. Nevertheless, during that time it may favorably influence the bradykinesia, rigidity, and tremor of parkinsonism. The standard dosage is 100 mg orally two or three times daily. Amantadine may also help in reducing iatrogenic dyskinesias in patients with advanced disease.

Adverse Effects Amantadine has a number of undesirable central nervous system effects, all of which can be reversed by stopping the drug. These include restlessness, depression, irritability, insomnia, agitation, excitement, hallucinations, and confusion. Overdosage may produce an acute toxic psychosis. With doses several times higher than recommended, convulsions have occurred. Livedo reticularis sometimes occurs in patients taking amantadine and usually clears within 1 month after the drug is withdrawn. Other dermatologic reactions have also been described. Peripheral edema, another well-recognized complication, is not accompanied by signs of cardiac, hepatic, or renal disease and responds to diuretics. Other adverse reactions to amantadine include headache, heart failure, postural hypotension, urinary retention, and gastrointestinal disturbances (eg, anorexia, nausea, constipation, and dry mouth). Amantadine should be used with caution in patients with a history of seizures or heart failure.

ACETYLCHOLINE-BLOCKING DRUGS A number of centrally acting antimuscarinic preparations are available that differ in their potency and in their efficacy in different patients. Some of these drugs were discussed in Chapter 8. These agents may improve the tremor and rigidity of parkinsonism but have little effect on bradykinesia. Some of the more commonly used drugs are listed in Table 28–1. TABLE 28–1 Some drugs with antimuscarinic properties used in parkinsonism.


Clinical Use Treatment is started with a low dose of one of the drugs in this category, the dosage gradually being increased until benefit occurs or until adverse effects limit further increments. If patients do not respond to one drug, a trial with another member of the drug class is warranted and may be successful.

Adverse Effects Antimuscarinic drugs have a number of undesirable central nervous system and peripheral effects (see Chapter 8) and are poorly tolerated by the elderly or cognitively impaired. Dyskinesias occur in rare cases. Acute suppurative parotitis sometimes occurs as a complication of dryness of the mouth. If medication is to be withdrawn, this should be accomplished gradually rather than abruptly to prevent acute exacerbation of parkinsonism. For contraindications to the use of antimuscarinic drugs, see Chapter 8.

SURGICAL PROCEDURES In patients with advanced disease that is poorly responsive to pharmacotherapy, worthwhile benefit may follow thalamotomy (for conspicuous tremor) or posteroventral pallidotomy. Ablative surgical procedures, however, have generally been replaced by functional, reversible lesions induced by high-frequency deep brain stimulation, which has a lower morbidity. Stimulation of the subthalamic nucleus or globus pallidus by an implanted electrode and stimulator has yielded good results for the management of the clinical fluctuations occurring in advanced parkinsonism. The anatomic substrate for such therapy is indicated in Figure 28–1. Such procedures are contraindicated in patients with secondary or atypical parkinsonism, dementia, or failure to respond to dopaminergic medication. The level of antiparkinsonian medication can often be reduced in patients undergoing deep brain stimulation, and this may help to ameliorate dose-related side effects of medication. In a controlled trial of the transplantation of dopaminergic tissue (fetal substantia nigra tissue), symptomatic benefit occurred in younger (less than 60 years old) but not older parkinsonian patients. In another trial, benefits were inconsequential. Furthermore, uncontrollable dyskinesias occurred in some patients in both studies, perhaps from a relative excess of dopamine from continued fiber outgrowth from the transplant. Additional basic studies are required before further trials of cellular therapies—in particular, stem cell therapies—are undertaken, and such approaches therefore remain investigational.

NEUROPROTECTIVE THERAPY Among the compounds under investigation as potential neuroprotective agents that may slow disease progression are antioxidants, antiapoptotic agents, glutamate antagonists, intraparenchymally administered glial-derived neurotrophic factor, and anti-inflammatory drugs. The role of these agents remains to be established, however, and their use for therapeutic purposes is not indicated at this time. Coenzyme Q10 and creatine have not been found effective despite early hopes to the contrary. The possibility that rasagiline has a protective effect was discussed earlier.

GENE THERAPY


Several phase 1 (safety) or phase 2 trials of gene therapy for Parkinson’s disease have been completed in the USA. All trials involved infusion into the striatum of adeno-associated virus type 2 as the vector for the gene. The genes were for glutamic acid decarboxylase (GAD, to facilitate synthesis of GABA, an inhibitory neurotransmitter), infused into the subthalamic nucleus to cause inhibition; for aromatic acid decarboxylase (AADC), infused into the putamen to increase metabolism of levodopa to dopamine; and for neurturin (a growth factor that may enhance the survival of dopaminergic neurons), infused into the putamen. All agents were deemed safe and the data suggested efficacy. A phase 2 study of the GAD gene has been completed and the results are encouraging, but one for neurturin infused into the substantia nigra as well as the putamen was disappointing. A phase 2 trial of AADC is planned.

THERAPY FOR NON-MOTOR MANIFESTATIONS Persons with cognitive decline may respond to rivastigmine (1.5–6 mg twice daily), memantine (5–10 mg daily), or donepezil (5–10 mg daily) (see Chapter 60); affective disorders to antidepressants or anxiolytic agents (see Chapter 30); excessive daytime sleepiness to modafinil (100–400 mg in the morning) (see Chapter 9); and bladder and bowel disorders to appropriate symptomatic therapy (see Chapter 8).

GENERAL COMMENTS ON DRUG MANAGEMENT OF PATIENTS WITH PARKINSONISM Parkinson’s disease generally follows a progressive course. Moreover, the benefits of levodopa therapy often diminish with time, and serious adverse effects may complicate long-term levodopa treatment. Nevertheless, dopaminergic therapy at a relatively early stage may be most effective in alleviating symptoms of parkinsonism and may also favorably affect the mortality rate due to the disease. Therefore, several strategies have evolved for optimizing dopaminergic therapy, as summarized in Figure 28–5. Symptomatic treatment of mild parkinsonism is probably best avoided until there is some degree of disability or until symptoms begin to impact the patient’s lifestyle. When symptomatic treatment becomes necessary, a trial of rasagiline, amantadine, or an antimuscarinic drug (in young patients) may be worthwhile. With disease progression, dopaminergic therapy becomes necessary. This can conveniently be initiated with a dopamine agonist, either alone or in combination with low-dose carbidopa-levodopa therapy, unless risk factors for impulse control disorders are present. Alternatively, especially in older patients, a dopamine agonist can be omitted and the patient started immediately on carbidopa-levodopa, which is the most effective symptomatic treatment of the motor disturbances of parkinsonism. Physical therapy is helpful in improving mobility. In patients with severe parkinsonism and long-term complications of levodopa therapy such as the on-off phenomenon, a trial of treatment with a COMT inhibitor or rasagiline may be helpful. Regulation of dietary protein intake may also improve response fluctuations. Deep brain stimulation is often helpful in patients who fail to respond adequately to these measures. Treating patients who are young or have mild parkinsonism with rasagiline may delay disease progression and merits consideration.

DRUG-INDUCED PARKINSONISM Reserpine and the related drug tetrabenazine deplete biogenic monoamines from their storage sites, whereas haloperidol, metoclopramide, and the phenothiazines block dopamine receptors. These drugs may therefore produce a parkinsonian syndrome, usually within 3 months after introduction. The disorder tends to be symmetric, with inconspicuous tremor, but this is not always the case. The syndrome is related to high dosage and clears over several weeks or months after withdrawal. If treatment is necessary, antimuscarinic agents are preferred. Levodopa is of no help if neuroleptic drugs are continued and may in fact aggravate the mental disorder for which antipsychotic drugs were prescribed originally. In 1983, a drug-induced form of parkinsonism was discovered in individuals who attempted to synthesize and use a narcotic drug related to meperidine but actually synthesized and self-administered MPTP, as discussed in the Box: MPTP & Parkinsonism.

OTHER MOVEMENT DISORDERS Tremor Tremor consists of rhythmic oscillatory movements. Physiologic postural tremor, which is a normal phenomenon, is enhanced in amplitude by anxiety, fatigue, thyrotoxicosis, and intravenous epinephrine or isoproterenol. Propranolol reduces its amplitude and, if administered intra-arterially, prevents the response to isoproterenol in the perfused limb, presumably through some peripheral action. Certain drugs—especially the bronchodilators, valproate, tricyclic antidepressants, and lithium—may produce a dose-dependent exaggeration of the normal physiologic tremor that is reversed by discontinuing the drug. Although the tremor produced by sympathomimetics such as terbutaline (a bronchodilator) is blocked by propranolol, which antagonizes both β1 and β2 receptors, it is not


blocked by metoprolol, a β1 -selective antagonist; this suggests that such tremor is mediated mainly by the β2 receptors. Essential tremor is a postural tremor, sometimes familial with autosomal dominant inheritance, which is clinically similar to physiologic tremor. At least three gene loci ( ETM1 on 3q13, ETM2 on 2p24.1, and a locus on 6p23) have been described as have associations with various other mapped loci. Dysfunction of β1 receptors has been implicated in some instances, since the tremor may respond dramatically to standard doses of metoprolol as well as to propranolol. The tremor may involve the hands, head, voice, and— much less commonly—the legs. Patients may become functionally limited or socially withdrawn, quality of life is affected, and some patients report being seriously disabled by the tremor. The most useful therapeutic approach is with propranolol, but whether the response depends on a central or peripheral action is unclear. The pharmacokinetics, pharmacologic effects, and adverse reactions of propranolol are discussed in Chapter 10. Total daily doses of propranolol on the order of 120 mg or more (range, 60–320 mg) are usually required, divided into two doses; reported adverse effects have been few. Propranolol should be used with caution in patients with heart failure, heart block, asthma, depression, or hypoglycemia. Other adverse effects include fatigue, malaise, lightheadedness, and impotence. Patients can be instructed to take their own pulse and call the physician if significant bradycardia develops. Long-acting propranolol is also effective and is preferred by many patients because of its convenience. Some patients prefer to take a single dose of propranolol when they anticipate their tremor is likely to be exacerbated, for example, by social situations. Metoprolol is sometimes useful in treating tremor when patients have concomitant pulmonary disease that contraindicates use of propranolol. Primidone (an antiepileptic drug; see Chapter 24), in gradually increasing doses up to 250 mg three times daily, is also effective in providing symptomatic control in some cases. Patients with tremor are very sensitive to primidone and often cannot tolerate the doses used to treat seizures; they should be started on 50 mg once daily and the daily dose increased by 50 mg every 2 weeks depending on response.

MPTP & Parkinsonism Reports in the early 1980s of a rapidly progressive form of parkinsonism in young persons opened a new area of research in the etiology and treatment of parkinsonism. The initial report described apparently healthy young people who attempted to support their opioid habit with a meperidine analog synthesized by an amateur chemist. They unwittingly self-administered 1-methyl-4-phenyl1,2,3,6-tetrahydropyridine (MPTP) and subsequently developed a very severe form of parkinsonism. MPTP is a protoxin that is converted by monoamine oxidase B to N-methyl-4-phenylpyridinium (MPP +). MPP + is selectively taken up by cells in the substantia nigra through an active mechanism normally responsible for dopamine reuptake. MPP + inhibits mitochondrial complex I, thereby inhibiting oxidative phosphorylation. The interaction of MPP + with complex I probably leads to cell death and thus to striatal dopamine depletion and parkinsonism. Recognition of the effects of MPTP suggested that spontaneously occurring Parkinson’s disease may result from exposure to an environmental toxin that is similarly selective in its target. However, no such toxin has yet been identified. It also suggested a successful means of producing an experimental model of Parkinson’s disease in animals, especially nonhuman primates. This model is useful in the development of new antiparkinsonism drugs. Pretreatment of exposed animals with a monoamine oxidase B inhibitor such as selegiline prevents the conversion of MPTP to MPP + and thus protects against the occurrence of parkinsonism. This observation has provided one reason to believe that selegiline or rasagiline may retard the progression of Parkinson’s disease in humans. Topiramate, another antiepileptic drug, may also be helpful in a dose of 400 mg daily, built up gradually. Alprazolam (in doses up to 3 mg daily) or gabapentin (100–2400 mg/d; typically 1200 mg/d) is helpful in some patients. Gabapentin is associated with fewer side effects than primidone. Other patients are helped by intramuscular injections of botulinum toxin, but dose-dependent weakness may complicate symptomatic benefit. Thalamic stimulation by an implanted electrode and stimulator is often worthwhile in advanced cases refractory to pharmacotherapy. MRI-guided focused ultrasound thalamotomy showed promise in a recent trial. Diazepam, chlordiazepoxide, mephenesin, and antiparkinsonism agents have been advocated in the past but are generally worthless. Small quantities of alcohol may suppress essential tremor for a short time but should not be recommended as a treatment strategy because of possible behavioral and other complications of alcohol. Intention tremor is present during movement but not at rest; sometimes it occurs as a toxic manifestation of alcohol or drugs such as phenytoin. Withdrawal or reduction in dosage provides dramatic relief. There is no satisfactory pharmacologic treatment for intention tremor due to other neurologic disorders. Rest tremor is usually due to parkinsonism.

Huntington’s Disease Huntington’s disease is an autosomal dominant inherited disorder caused by an abnormality (expansion of a CAG trinucleotide repeat that


codes for a polyglutamine tract) of the huntingtin gene on chromosome 4. An autosomal recessive form may also occur. Huntington disease–like (HDL) disorders are not associated with an abnormal CAG trinucleotide repeat number of the huntingtin gene. Autosomal dominant (HDL1, 20pter-p12; HDL2, 16q24.3) and recessive forms (HDL3, 4p15.3) occur. Huntington’s disease is characterized by progressive chorea and dementia that usually begin in adulthood. The development of chorea seems to be related to an imbalance of dopamine, acetylcholine, GABA, and perhaps other neurotransmitters in the basal ganglia (Figure 28–6). Pharmacologic studies indicate that chorea results from functional overactivity in dopaminergic nigrostriatal pathways, perhaps because of increased responsiveness of post-synaptic dopamine receptors or deficiency of a neurotransmitter that normally antagonizes dopamine. Drugs that impair dopaminergic neurotransmission, either by depleting central monoamines (eg, reserpine, tetrabenazine) or by blocking dopamine receptors (eg, phenothiazines, butyrophenones), often alleviate chorea, whereas dopamine-like drugs such as levodopa tend to exacerbate it.

FIGURE 28–6 Schematic representation of the sequence of neurons involved in Huntington’s chorea. Top: Dopaminergic neurons (red) originating in the substantia nigra normally inhibit the GABAergic output from the striatum, whereas cholinergic neurons (green) exert an excitatory effect. Bottom: In Huntington’s chorea, some cholinergic neurons may be lost, but even more GABAergic neurons (black) degenerate. Both GABA and the enzyme (glutamic acid decarboxylase) concerned with its synthesis are markedly reduced in the basal ganglia of patients with Huntington’s disease, and GABA receptors are usually implicated in inhibitory pathways. There is also a significant decline in concentration of choline acetyltransferase, the enzyme responsible for synthesizing acetylcholine, in the basal ganglia of these patients. These findings may be of pathophysiologic significance and have led to attempts to alleviate chorea by enhancing central GABA or acetylcholine activity, but with disappointing results. As a consequence, the most commonly used drugs for controlling dyskinesia in patients with Huntington’s disease are still those that interfere with dopamine activity. With all the latter drugs, however, reduction of abnormal movements may be associated with iatrogenic parkinsonism. Tetrabenazine (12.5–50 mg orally three times daily) depletes cerebral dopamine and reduces the severity of chorea. It has less troublesome adverse effects than reserpine, which has also been used for this purpose. Tetrabenazine is metabolized by cytochrome P450 (CYP2D6), and genotyping has therefore been recommended to determine metabolizer status (CYP2D6 expression) in patients needing doses exceeding 50 mg/d. For poor metabolizers, the maximum recommended dose is 50 mg daily (25 mg/dose); otherwise, a maximum dose of 100 mg daily can be used. Treatment with postsynaptic dopamine receptor blockers such as phenothiazines and butyrophenones may also be helpful. Haloperidol is started in a small dose, eg, 1 mg twice daily, and increased every 4 days depending on the response. If haloperidol is not helpful, treatment with increasing doses of fluphenazine in a similar dose, eg, 1 mg twice daily, sometimes helps. Several recent reports suggest that olanzapine may also be useful; the dose varies with the patient, but 10 mg daily is often sufficient, although doses as high as 30 mg daily are sometimes required. The pharmacokinetics and clinical properties of these drugs are considered in greater detail elsewhere in this book. Selective serotonin reuptake inhibitors may reduce depression, aggression, and agitation. However, strong CYP2D6 inhibitors should be used with caution, as it may be necessary to decrease the dose of


tetrabenazine taken concurrently. Other important aspects of management include genetic counseling, speech therapy, physical and occupational therapy, dysphagia precautions, and provision of social services.

Other Forms of Chorea Benign hereditary chorea is inherited (usually autosomal dominant; possibly also autosomal recessive) or arises spontaneously. Chorea develops in early childhood and does not progress during adult life; dementia does not occur. In patients with TITF-1 gene mutations, thyroid and pulmonary abnormalities may also be present (brain-thyroid-lung syndrome). Familial chorea may also occur as part of the chorea-acanthocytosis syndrome, together with orolingual tics, vocalizations, cognitive changes, seizures, peripheral neuropathy, and muscle atrophy; serum β-lipoproteins are normal. Mutations of the gene encoding chorein at 9q21 may be causal. Treatment of these hereditary disorders is symptomatic. Tetrabenazine (0.5 mg/kg/d for children and 37.5 mg/d for adults) may improve chorea in some instances. Treatment is directed at the underlying cause when chorea occurs as a complication of general medical disorders such as thyrotoxicosis, polycythemia vera rubra, systemic lupus erythematosus, hypocalcemia, and hepatic cirrhosis. Drug-induced chorea is managed by withdrawal of the offending substance, which may be levodopa, an antimuscarinic drug, amphetamine, lithium, phenytoin, or an oral contraceptive. Neuroleptic drugs may also produce an acute or tardive dyskinesia (discussed below). Sydenham’s chorea is temporary and usually so mild that pharmacologic management of the dyskinesia is unnecessary, but dopamine-blocking drugs are effective in suppressing it.

Ballismus The biochemical basis of ballismus is unknown, but the pharmacologic approach to management is the same as for chorea. Treatment with tetrabenazine, haloperidol, perphenazine, or other dopamine-blocking drugs may be helpful.

Athetosis & Dystonia The pharmacologic basis of these disorders is unknown, and there is no satisfactory medical treatment for them. A subset of patients respond well to levodopa medication (dopa-responsive dystonia), which is therefore worthy of trial. Occasional patients with dystonia may respond to diazepam, amantadine, antimuscarinic drugs (in high dosage), carbamazepine, baclofen, haloperidol, or phenothiazines. A trial of these pharmacologic approaches is worthwhile, though often not successful. Patients with focal dystonias such as blepharospasm or torticollis often benefit from injection of botulinum toxin into the overactive muscles. Deep brain stimulation may be helpful in medically intractable cases.

Tics The pathophysiologic basis of tics is unknown. Chronic multiple tics (Gilles de la Tourette’s syndrome ) may require symptomatic treatment if the disorder is severe or is having a significant impact on the patient’s life. Education of patients, family, and teachers is important. Pharmacologic therapy may be necessary when tics interfere with social life or otherwise impair activities of daily living. Treatment is with drugs that block dopamine receptors or deplete dopamine stores, such as fluphenazine, pimozide, and tetrabenazine. These drugs reduce the frequency and intensity of tics by about 60%. Pimozide, a dopamine receptor antagonist, may be helpful in patients as a first-line treatment or in those who are either unresponsive to or intolerant of the other agents mentioned. Treatment is started at 1 mg/d, and the dosage is increased by 1 mg every 5 days; most patients require 7–16 mg/d. It has similar side effects to haloperidol but may cause irregularities of cardiac rhythm. Haloperidol has been used for many years to treat tic disorders. Patients are better able to tolerate this drug if treatment is started with a small dosage (eg, 0.25 or 0.5 mg daily) and then increased gradually (eg, by 0.25 mg every 4 or 5 days) over the following weeks depending on response and tolerance. Most patients ultimately require a total daily dose of 3–8 mg. Adverse effects include extrapyramidal movement disorders, sedation, dryness of the mouth, blurred vision, and gastrointestinal disturbances. Aripiprazole (see Chapter 29) has also been found effective in treating tics. Although not approved by the FDA for the treatment of tics or Tourette’s syndrome, certain α2 -adrenergic agonists may be preferred as an initial treatment because they are less likely to cause extrapyramidal side effects than neuroleptic agents. Clonidine reduces motor or vocal tics in about 50% of children so treated. It may act by reducing activity in noradrenergic neurons in the locus caeruleus. It is introduced at a dose of 2–3 mcg/kg/d, increasing after 2 weeks to 4 mcg/kg/d and then, if required, to 5 mcg/kg/d. It may cause an initial transient fall in blood pressure. The most common adverse effect is sedation; other adverse effects include reduced or excessive salivation and diarrhea. Guanfacine, another α2 -adrenergic agonist, has also been used. Both of these drugs may be particularly helpful for behavioral symptoms, such as impulse control disorders. Atypical antipsychotics, such as risperidone and aripiprazole, may be especially worthwhile in patients with significant behavioral problems. Clonazepam and carbamazepine have also been used. The pharmacologic properties of these drugs are discussed elsewhere in


this book. Injection of botulinum toxin A at the site of problematic tics is sometimes helpful when these are focal simple tics. Treatment of any associated attention deficit disorder (eg, with clonidine patch, guanfacine, pemoline, methylphenidate, or dextroamphetamine) or obsessive-compulsive disorder (with selective serotonin reuptake inhibitors or clomipramine) may be required. Deep brain stimulation is sometimes worthwhile in otherwise intractable cases but is best regarded as an investigational approach at this time.

Drug-Induced Dyskinesias Levodopa or dopamine agonists produce diverse dyskinesias as a dose-related phenomenon in patients with Parkinson’s disease; dose reduction reverses them. Chorea may also develop in patients receiving phenytoin, carbamazepine, amphetamines, lithium, and oral contraceptives, and it resolves with discontinuance of the offending medication. Dystonia has resulted from administration of dopaminergic agents, lithium, serotonin reuptake inhibitors, carbamazepine, and metoclopramide; and postural tremor from theophylline, caffeine, lithium, valproic acid, thyroid hormone, tricyclic antidepressants, and isoproterenol. The pharmacologic basis of the acute dyskinesia or dystonia sometimes precipitated by the first few doses of a phenothiazine is not clear. In most instances, parenteral administration of an antimuscarinic drug such as benztropine (2 mg intravenously), diphenhydramine (50 mg intravenously), or biperiden (2–5 mg intravenously or intramuscularly) is helpful, whereas in other instances diazepam (10 mg intravenously) alleviates the abnormal movements. Tardive dyskinesia, a disorder characterized by a variety of abnormal movements, is a common complication of long-term neuroleptic or metoclopramide drug treatment (see Chapter 29). Its precise pharmacologic basis is unclear. A reduction in dose of the offending medication, a dopamine receptor blocker, commonly worsens the dyskinesia, whereas an increase in dose may suppress it. The drugs most likely to provide immediate symptomatic benefit are those interfering with dopaminergic function, either by depletion (eg, reserpine, tetrabenazine) or receptor blockade (eg, phenothiazines, butyrophenones). Paradoxically, the receptor-blocking drugs are the very ones that also cause the dyskinesia. Tardive dystonia is usually segmental or focal; generalized dystonia is less common and occurs in younger patients. Treatment is the same as for tardive dyskinesia, but anticholinergic drugs may also be helpful; focal dystonias may also respond to local injection of botulinum A toxin. Tardive akathisia is treated similarly to drug-induced parkinsonism. Rabbit syndrome, another neuroleptic-induced disorder, is manifested by rhythmic vertical movements about the mouth; it may respond to anticholinergic drugs. Because the tardive syndromes that develop in adults are often irreversible and have no satisfactory treatment, care must be taken to reduce the likelihood of their occurrence. Antipsychotic medication should be prescribed only when necessary and should be withheld periodically to assess the need for continued treatment and to unmask incipient dyskinesia. Thioridazine, a phenothiazine with a piperidine side chain, is an effective antipsychotic agent that seems less likely than most to cause extrapyramidal reactions, perhaps because it has little effect on dopamine receptors in the striatal system. Finally, antimuscarinic drugs should not be prescribed routinely in patients receiving neuroleptics, because the combination may increase the likelihood of dyskinesia. Neuroleptic malignant syndrome, a rare complication of treatment with neuroleptics, is characterized by rigidity, fever, changes in mental status, and autonomic dysfunction (see Table 16–4). Symptoms typically develop over 1–3 days (rather than minutes to hours as in malignant hyperthermia) and may occur at any time during treatment. Treatment includes withdrawal of antipsychotic drugs, lithium, and anticholinergics; reduction of body temperature; and rehydration. Dantrolene, dopamine agonists, levodopa, or amantadine may be helpful, but there is a high mortality rate (up to 20%) with neuroleptic malignant syndrome.

Restless Legs Syndrome Restless legs syndrome is characterized by an unpleasant creeping discomfort that seems to arise deep within the legs and occasionally the arms. Symptoms occur particularly when patients are relaxed, especially when they are lying down or sitting, and they lead to an urge to move about. Such symptoms may delay the onset of sleep. A sleep disorder associated with periodic movements during sleep may also occur. The cause is unknown, but the disorder is especially common among pregnant women and also among uremic or diabetic patients with neuropathy. In most patients, no obvious predisposing cause is found, but several genetic loci have been associated with it. Symptoms may resolve with correction of coexisting iron-deficiency anemia and often respond to dopamine agonists, levodopa, diazepam, clonazepam, gabapentin, or opiates. Dopaminergic therapy is the preferred treatment for restless legs syndrome and should be initiated with long-acting dopamine agonists (eg, pramipexole 0.125–0.75 mg or ropinirole 0.25–4.0 mg once daily) or with the rotigotine skin patch to avoid the augmentation that may be associated, especially with levodopa-carbidopa (100/25 or 200/50 taken about 1 hour before bedtime). Augmentation refers to the earlier onset or enhancement of symptoms; earlier onset of symptoms at rest; and a briefer response to medication. When augmentation occurs with levodopa, a dopamine agonist should be substituted. If it occurs in patients receiving an agonist, the daily dose should be divided, another agonist tried, or other medications substituted. Dopamine agonist therapy may be associated with development of impulse control disorders. Gabapentin is effective in reducing the severity of restless legs syndrome and is taken once or twice daily (in the evening and before sleep). The starting dose is 300 mg daily, building up depending on response and tolerance (to approximately 1800 mg daily). Oral gabapentin enacarbil (600 or 1200 mg once daily) may also be helpful. A recent study suggests that pregabalin, a related drug, is also effective at a daily total dosage of 150–300 mg, taken in divided doses.


Clonazepam, 1 mg daily, is also sometimes helpful, especially for those with intermittent symptoms. When opiates are required, those with long half-lives or low addictive potential should be used. Oxycodone is often effective; the dose is individualized.

Wilson’s Disease A recessively inherited (13q14.3–q21.1) disorder of copper metabolism, Wilson’s disease is characterized biochemically by reduced serum copper and ceruloplasmin concentrations, pathologically by markedly increased concentration of copper in the brain and viscera, and clinically by signs of hepatic and neurologic dysfunction. Neurologic signs include tremor, choreiform movements, rigidity, hypokinesia, and dysarthria and dysphagia. Siblings of affected patients should be screened for asymptomatic Wilson’s disease. Treatment involves the removal of excess copper, followed by maintenance of copper balance. Dietary copper should also be kept below 2 mg daily. Penicillamine (dimethylcysteine) has been used for many years as the primary agent to remove copper. It is a chelating agent that forms a ring complex with copper. It is readily absorbed from the gastrointestinal tract and rapidly excreted in the urine. A common starting dose in adults is 500 mg three or four times daily. After remission occurs, it may be possible to lower the maintenance dose, generally to not less than 1 g daily, which must thereafter be continued indefinitely. Adverse effects include nausea and vomiting, nephrotic syndrome, a lupus-like syndrome, pemphigus, myasthenia, arthropathy, optic neuropathy, and various blood dyscrasias. In about 10% of instances, neurologic worsening occurs with penicillamine. Treatment should be monitored by frequent urinalysis and complete blood counts. Trientine hydrochloride, another chelating agent, is preferred by many over penicillamine because of the lesser likelihood of drug reactions or neurologic worsening. It may be used in a daily dose of 1–1.5 g. Trientine appears to have few adverse effects other than mild anemia due to iron deficiency in a few patients. Tetrathiomolybdate may be better than trientine for preserving neurologic function in patients with neurologic involvement and is taken both with and between meals. It is not yet commercially available. Zinc acetate administered orally increases the fecal excretion of copper and can be used in combination with these other agents. The dose is 50 mg three times a day. Zinc sulfate (200 mg/d orally) has also been used to decrease copper absorption. Zinc blocks copper absorption from the gastrointestinal tract by induction of intestinal cell metallothionein. Its main advantage is its low toxicity compared with that of other anticopper agents, although it may cause gastric irritation when introduced. Liver transplantation is sometimes necessary. The role of hepatocyte transplantation and gene therapy is currently under investigation.

SUMMARY Drugs Used for Movement Disorders



PREPARATIONS AVAILABLE


REFERENCES Allain H et al: Disease-modifying drugs and Parkinson’s disease. Prog Neurobiol 2008;84:25. Aminoff MJ: T reatment should not be initiated too soon in Parkinson’s disease. Ann Neurol 2006;59:562. Angot E et al: Are synucleinopathies prion-like disorders? Lancet Neurol 2010;9:1128. Antonini A et al: Role of pramipexole in the management of Parkinson’s disease. CNS Drugs 2010;24:829. Bestha DP et al: Management of tics and T ourette’s disorder: An update. Expert Opin Pharmacother 2010;11:1813. Brewer GJ: T he use of copper-lowering therapy with tetrathiomolybdate in medicine. Expert Opin Investig Drugs 2009;18:89. Bronstein JM et al: Deep brain stimulation for Parkinson disease: An expert consensus and review of key issues. Arch Neurol 2010;68:165. Christine CW et al: Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology 2009;73:1662. Cornelius JR et al: Impulse control disorders with the use of dopaminergic agents in restless legs syndrome: A case control study. Sleep 2010;33:81. Earley CJ et al: Restless legs syndrome and periodic leg movements in sleep. Handb Clin Neurol 2011;99:913. Elias WJ et al: A pilot study of focused ultrasound thalamotomy for essential tremor. N Engl J Med 2013;369:640. Evans AH et al: Impulsive and compulsive behaviors in Parkinson’s disease. Mov Disord 2009;24:1561. Follett KA et al: Pallidal versus subthalamic deep-brain stimulation for Parkinson’s disease. N Engl J Med 2010;362:2077. Frank S: T etrabenazine as anti-chorea therapy in Huntington disease: An open-label continuation study. Huntington Study Group/T ET RA-HD Investigators. BMC Neurol 2009;9:62. Frank S, Jankovic J: Advances in the pharmacological management of Huntington’s disease. Drugs 2010;70:561. Garcia-Borreguero D et al: T reatment of restless legs syndrome with pregabalin: A double-blind, placebo-controlled study. Neurology 2010;74:1897. Gottwald MD, Aminoff MJ: T herapies for dopaminergic-induced dyskinesias in Parkinson disease. Ann Neurol 2011;69:919. Hartmann A, Worbe Y: Pharmacological treatment of Gilles de la T ourette syndrome. Neurosci Biobehav Rev 2013;37:1157. Hauser RA et al: Double-blind trial of levodopa/carbidopa/entacapone versus levodopa/carbidopa in early Parkinson’s disease. Mov Disord 2009;24:541. Huster D: Wilson disease. Best Pract Res Clin Gastroenterol 2010;24:531. Jankovic J: Medical treatment of dystonia. Mov Disord 2013;28:1001. Kimber T E: An update on T ourette syndrome. Curr Neurol Neurosci Rep 2010;10:286. Kordower JH et al: T ransplanted dopaminergic neurons develop PD pathologic changes: A second case report. Mov Disord 2008;23:2303. LeWitt PA et al: AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol 2011;10:309. Lorincz MT : Neurologic Wilson’s disease. Ann N Y Acad Sci 2010;1184:173. Lyons KE, Pahwa R: Outcomes of rotigotine clinical trials: Effects on motor and nonmotor symptoms of Parkinson’s disease. Neurol Clin 2013;31(3 Suppl):S51. Marks WJ Jr et al: Gene delivery of AAV2-neurturin for Parkinson’s disease: A double-blind, randomised, controlled trial. Lancet Neurol 2010;9:1164. Olanow CW et al: A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009;361:1268. Perez-Lloret et al: Adverse drug reactions to dopamine agonists: a comparative study in the French Pharmacovigilance Database. Mov Disord 2010;25:1876. Sadeghi R, Ondo WG: Pharmacological management of essential tremor. Drugs 2010;70:2215. Schilsky ML: Wilson disease: Current status and the future. Biochimie 2009;91:1278. Scott LJ: Gabapentin enacarbil: In patients with restless legs syndrome. CNS Drugs 2012;26:1073. Seeberger LC, Hauser RA: Levodopa/carbidopa/entacapone in Parkinson’s disease. Expert Rev Neurother 2009;9:929. Servello D et al: Deep brain stimulation in 18 patients with severe Gilles de la T ourette syndrome refractory to treatment: T he surgery and stimulation. J Neurol Neurosurg Psychiatry 2008;79:136. Singer HS: T reatment of tics and T ourette syndrome. Curr T reat Options Neurol 2010;12:539. Stocchi F et al: Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: T he ST RIDE-PD study. Ann Neurol 2010;68:18. T amara P et al: T ourette syndrome and other tic disorders of childhood. Handb Clin Neurol 2013;112:853. T aylor RM et al: T riethylene tetramine dihydrochloride (trientine) in children with Wilson disease: Experience at King’s College Hospital and review of the literature. Eur J Pediatr 2009;168:1061. T homas R, Cavanna AE: T he pharmacology of T ourette syndrome. J Neural T ransm 2013;120:689. T renkwalder C, Paulus W: Restless legs syndrome: Pathophysiology, clinical presentation and management. Nat Rev Neurol 2010;6:337. Van Holst et al: Brain imaging studies in pathological gambling. Curr Psychiatry Rep 2010;12:418. Videnovic A: T reatment of Huntington disease. Curr T reat Options Neurol 2013;15:424. Vilas D et al: Impulse control disorders in Parkinson’s disease. Parkinsonism Relat Disord 2012;18, Suppl 1:S80. Voon V et al: Impulse control disorders in Parkinson’s disease: A multicenter case-control study. Ann Neurol 2011;69:986. Weaver FM et al: Randomized trial of deep brain stimulation for Parkinson disease: T hirty-six-month outcomes. Neurology 2012;79:55. Weintraub D et al: Impulse control disorders in Parkinson disease: A cross-sectional study of 3090 patients. Arch Neurol 2010;67:589. Welter ML et al: Internal pallidal and thalamic stimulation in patients with T ourette syndrome. Arch Neurol 2008; 65:952. Wiggelinkhuizen M et al: Systematic review: Clinical efficacy of chelator agents and zinc in the initial treatment of Wilson disease. Aliment Pharmacol T her 2009;29:947. Zesiewicz T A et al: Update on treatment of essential tremor. Curr T reat Options Neurol 2013;15:410.

CASE STUDY ANSWER The relation of the tremor to activity (rest tremor) in this case is characteristic of parkinsonism. Examination reveals the classic findings of Parkinson’s disease—rest tremor, rigidity, bradykinesia, and a gait disturbance; an asymmetry of the abnormalities is common in Parkinson’s disease. The prognosis is that symptoms will become more generalized with time. Pharmacologic treatment would involve a dopamine agonist (pramipexole or ropinirole) but may not need to be started now unless the patient is disturbed by


his symptoms. The patient developed an impulse control disorder (gambling) after starting on an agonist, and this may require dose reduction or discontinuation of the agonist.


CHAPTER

29 Antipsychotic Agents & Lithium Charles DeBattista, MD*

CASE STUDY A 17-year-old male high school student is referred to the psychiatry clinic for evaluation of suspected schizophrenia. After a diagnosis is made, haloperidol is prescribed at a gradually increasing dose on an outpatient basis. The drug improves the patient’s positive symptoms but ultimately causes intolerable adverse effects. Although more costly, risperidone is then prescribed, which, over the course of several weeks of treatment, improves his symptoms and is tolerated by the patient. What signs and symptoms would support an initial diagnosis of schizophrenia? In the treatment of schizophrenia, what benefits do the atypical antipsychotic drugs offer over the traditional agents such as haloperidol? In addition to the management of schizophrenia, what other clinical indications warrant consideration of the use of drugs nominally classified as antipsychotics?

ANTIPSYCHOTIC AGENTS Antipsychotic drugs are able to reduce psychotic symptoms in a wide variety of conditions, including schizophrenia, bipolar disorder, psychotic depression, senile psychoses, various organic psychoses, and drug-induced psychoses. They are also able to improve mood and reduce anxiety and sleep disturbances, but they are not the treatment of choice when these symptoms are the primary disturbance in nonpsychotic patients. A neuroleptic is a subtype of antipsychotic drug that produces a high incidence of extrapyramidal side effects (EPS) at clinically effective doses, or catalepsy in laboratory animals. The “atypical” antipsychotic drugs are now the most widely used type of antipsychotic drug.

History Reserpine and chlorpromazine were the first drugs found to be useful to reduce psychotic symptoms in schizophrenia. Reserpine was used only briefly for this purpose and is no longer of interest as an antipsychotic agent. Chlorpromazine is a neuroleptic agent; that is, it produces catalepsy in rodents and EPS in humans. The discovery that its antipsychotic action was related to dopamine (D or DA)receptor blockade led to the identification of other compounds as antipsychotics between the 1950s and 1970s. The discovery of clozapine in 1959 led to the realization that antipsychotic drugs need not cause EPS in humans at clinically effective doses. Clozapine was called an atypical antipsychotic drug because of this dissociation; it produces fewer EPS at equivalent antipsychotic doses in man and laboratory animals. As a result, there has been a major shift in clinical practice away from typical antipsychotic drugs towards the use of an ever increasing number of atypical drugs, which have other advantages as well. The introduction of antipsychotic drugs led to massive changes in disease management, including brief instead of life-long hospitalizations. These drugs have also proved to be of great value in studying the pathophysiology of schizophrenia and other psychoses. It should be noted that schizophrenia and bipolar disorder are no longer believed by many to be separate disorders but rather to be part of a continuum of brain disorders with psychotic features.

Nature of Psychosis & Schizophrenia The term “psychosis” denotes a variety of mental disorders: the presence of delusions (false beliefs), various types of hallucinations, usually auditory or visual, but sometimes tactile or olfactory, and grossly disorganized thinking in a clear sensorium. Schizophrenia is a particular kind of psychosis characterized mainly by a clear sensorium but a marked thinking disturbance. Psychosis is not unique to schizophrenia and is not present in all patients with schizophrenia at all times. Schizophrenia is considered to be a neurodevelopmental disorder. This implies that structural and functional changes in the brain are present even in utero in some patients, or that they develop during childhood and adolescence, or both. Twin, adoption, and family studies


have established that schizophrenia is a genetic disorder with high heritability. No single gene is involved. Current theories involve multiple genes with common and rare mutations, including large deletions and insertions (copy number variations), combining to produce a very variegated clinical presentation and course.

THE SEROTONIN HYPOTHESIS OF SCHIZOPHRENIA The discovery that indole hallucinogens such as LSD (lysergic acid diethylamide) and mescaline are serotonin (5-HT) agonists led to the search for endogenous hallucinogens in the urine, blood, and brains of patients with schizophrenia. This proved fruitless, but the identification of many 5-HT-receptor subtypes led to the pivotal discovery that 5-HT 2A-receptor and possibly 5-HT2C stimulation was the basis for the hallucinatory effects of these agents. It has been found that 5-HT2A-receptor blockade is a key factor in the mechanism of action of the main class of atypical antipsychotic drugs, of which clozapine is the prototype, and includes, in order of their introduction around the world, melperone, risperidone, zotepine, blonanserin, olanzapine, quetiapine, ziprasidone, aripiprazole, sertindole, paliperidone, iloperidone, asenapine, and lurasidone. These drugs are inverse agonists of the 5-HT2A receptor; that is, they block the constitutive activity of these receptors. These receptors modulate the release of dopamine, norepinephrine, glutamate, GABA, and acetylcholine, among other neurotransmiters in the cortex, limbic region, and striatum. Stimulation of 5-HT2A receptors leads to depolarization of glutamate neurons, but also stabilization of N-methyl-D-aspartate (NMDA) receptors on postsynaptic neurons. Recently, it has been found that hallucinogens can modulate the stability of a complex consisting of 5-HT2A and NMDA receptors. 5-HT2C-receptor stimulation provides a further means of modulating cortical and limbic dopaminergic activity. Stimulation of 5-HT 2C receptors leads to inhibition of cortical and limbic dopamine release. Many atypical antipsychotic drugs, eg, clozapine, asenapine, olanzapine, are 5-HT2C inverse agonists. 5-HT2C agonists are currently being studied as antipsychotic agents.

THE DOPAMINE HYPOTHESIS OF SCHIZOPHRENIA The dopamine hypothesis for schizophrenia was the second neurotransmitter-based concept to be developed but is no longer considered adequate to explain all aspects of schizophrenia, especially the cognitive impairment. Nevertheless, it is still highly relevant to understanding the major dimensions of schizophrenia, such as positive and negative symptoms (emotional blunting, social withdrawal, lack of motivation), cognitive impairment, and possibly depression. It is also essential to understanding the mechanisms of action of most and probably all antipsychotic drugs. Several lines of evidence suggest that excessive limbic dopaminergic activity plays a role in psychosis. (1) Many antipsychotic drugs strongly block postsynaptic D2 receptors in the central nervous system, especially in the mesolimbic and striatal-frontal system; this includes partial dopamine agonists, such as aripiprazole and bifeprunox. (2) Drugs that increase dopaminergic activity, such as levodopa, amphetamines, and bromocriptine and apomorphine, either aggravate schizophrenia psychosis or produce psychosis de novo in some patients. (3) Dopamine-receptor density has been found postmortem to be increased in the brains of schizophrenics who have not been treated with antipsychotic drugs. (4) Some but not all postmortem studies of schizophrenic subjects have reported increased dopamine levels and D2 -receptor density in the nucleus accumbens, caudate, and putamen. (5) Imaging studies have shown increased amphetamine-induced striatal dopamine release, increased baseline occupancy of striatal D2 receptors by extracellular dopamine, and other measures consistent with increased striatal dopamine synthesis and release. However, the dopamine hypothesis is far from a complete explanation of all aspects of schizophrenia. Diminished cortical or hippocampal dopaminergic activity has been suggested to underlie the cognitive impairment and negative symptoms of schizophrenia. Postmortem and in vivo imaging studies of cortical, limbic, nigral, and striatal dopaminergic neurotransmission in schizophrenic subjects have reported findings consistent with diminished dopaminergic activity in these regions. Decreased dopaminergic innervation in medial temporal cortex, dorsolateral prefrontal cortex, and hippocampus, and decreased levels of DOPAC, a metabolite of dopamine, in the anterior cingulate have been reported in postmortem studies. Imaging studies have found increased prefrontal D1 -receptor levels that correlated with working memory impairments. The fact that several of the atypical antipsychotic drugs have much less effect on D2 receptors and yet are effective in schizophrenia has redirected attention to the role of other dopamine receptors and to nondopamine receptors. Serotonin receptors—particularly the 5HT2A-receptor subtype—may mediate synergistic effects or protect against the extrapyramidal consequences of D2 antagonism. As a result of these considerations, the direction of research has changed to a greater focus on compounds that may act on several transmitter-receptor systems, eg, serotonin and glutamate. The atypical antipsychotic drugs share the property of weak D2 -receptor antagonism and more potent 5-HT2A-receptor blockade.

THE GLUTAMATE HYPOTHESIS OF SCHIZOPHRENIA


Glutamate is the major excitatory neurotransmitter in the brain (see Chapter 21). Phencyclidine (PCP) and ketamine are noncompetitive inhibitors of the NMDA receptor that exacerbate both cognitive impairment and psychosis in patients with schizophrenia. PCP and a related drug, MK-801, increase locomotor activity and, acutely or chronically, a variety of cognitive impairments in rodents and primates. These effects are widely employed as a means to develop novel antipsychotic and cognitive-enhancing drugs. Selective 5-HT2A antagonists, as well as atypical antipsychotic drugs, are much more potent than D2 antagonists in blocking these effects of PCP and MK801. This was the starting point for the hypothesis that hypofunction of NMDA receptors, located on GABAergic interneurons, leading to diminished inhibitory influences on neuronal function, contributed to schizophrenia. The diminished GABAergic activity can induce disinhibition of downstream glutamatergic activity, which can lead to hyperstimulation of cortical neurons through non-NMDA receptors. Preliminary evidence suggests that LY2140023, a drug that acts as an agonist of the metabotropic 2/3 glutamate receptor (mGLuR2/3), may be effective in schizophrenia. The NMDA receptor, an ion channel, requires glycine for full activation. It has been suggested that in patients with schizophrenia, the glycine site of the NMDA receptor is not fully saturated. There have been several trials of high doses of glycine to promote glutamatergic activity, but the results are far from convincing. Currently, glycine transport inhibitors are in development as possible antipsychotic agents. Ampakines are drugs that potentiate currents mediated by AMPA-type glutamate receptors. In behavioral tests, ampakines are effective in correcting behaviors in various animal models of schizophrenia and depression. They protect neurons against neurotoxic insults, in part by mobilizing growth factors such as brain-derived neurotrophic factor (BDNF, see also Chapter 30).

BASIC PHARMACOLOGY OF ANTIPSYCHOTIC AGENTS Chemical Types A number of chemical structures have been associated with antipsychotic properties. The drugs can be classified into several groups as shown in Figures 29–1 and 29–2.


FIGURE 29–1 Structural formulas of some older antipsychotic drugs: phenothiazines, thioxanthenes, and butyrophenones. Only representative members of each type are shown.



FIGURE 29–2 Structural formulas of some newer antipsychotic drugs. A. Phenothiazine Derivatives Three subfamilies of phenothiazines, based primarily on the side chain of the molecule, were once the most widely used of the antipsychotic agents. Aliphatic derivatives (eg, chlorpromazine) and piperidine derivatives (eg, thioridazine) are the least potent. These drugs produce more sedation and weight gain. Piperazine derivatives are more potent (effective in lower doses) but not necessarily more efficacious. The piperazine derivatives are also more selective in their pharmacologic effects (Table 29–1). TABLE 29–1 Antipsychotic drugs: Relation of chemical structure to potency and toxicities.

The National Institute of Mental Health (NIMH)-funded Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) reported that perphenazine, a piperazine derivative, was as effective as atypical antipsychotic drugs, with the modest exception of olanzapine, and concluded that typical antipsychotic drugs are the treatment of choice for schizophrenia based on their lower cost. However, there were numerous flaws in the design, execution and analysis of this study, leading to it having only modest impact on clinical practice. In particular, it failed to consider issues such as dosage of olanzapine, inclusion of treatment resistant patients, encouragement of patients to switch medications inherent in the design, risk for tardive dyskinesia following long-term use of even low dose typical antipsychotics, and the necessity of large sample sizes in equivalency studies. B. Thioxanthene Derivatives This group of drugs is exemplified primarily by thiothixene.


C. Butyrophenone Derivatives This group, of which haloperidol is the most widely used, has a very different structure from those of the two preceding groups. Haloperidol, a butyrophenone, is the most widely used typical antipsychotic drug, despite its high level of EPS relative to typical antipsychotic drugs. Diphenylbutylpiperidines are closely related compounds. The butyrophenones and congeners tend to be more potent and to have fewer autonomic effects but greater extrapyramidal effects than phenothiazines (Table 29–1). D. Miscellaneous Structures Pimozide and molindone are typical antipsychotic drugs. There is no significant difference in efficacy between these newer typical and the older typical antipsychotic drugs. E. Atypical Antipsychotic Drugs Clozapine, asenapine, olanzapine, quetiapine, paliperidone, risperidone, sertindole, ziprasidone, zotepine, and aripiprazole are atypical antipsychotic drugs (some of which are shown in Figure 29–2). Clozapine is the prototype. Paliperidone is 9hydroxyrisperidone, the active metabolite of risperidone. Risperidone is rapidly converted to 9-hydroxyrisperidone in vivo in most patients, except for about 10% of patients who are poor metabolizers. Sertindole is approved in some European countries but not in the USA. These drugs have complex pharmacology but they share a greater ability to alter 5-HT2A-receptor activity than to interfere with D2 receptor action. In most cases, they act as partial agonists at the 5-HT1A receptor, which produces synergistic effects with 5-HT 2A receptor antagonism. Most are either 5-HT6 or 5-HT7 receptor antagonists. Sulpride and sulpiride constitute another class of atypical agents. They have equivalent potency for D2 and D3 receptors, but they are also 5-HT7 antagonists. They dissociate EPS and antipsychotic efficacy. However, they also produce marked increases in serum prolactin levels and are not as free of the risk of tardive dyskinesia as are drugs such as clozapine and quetiapine. They are not approved in the USA. Cariprazine represents another class of atypical agents. In addition to D2 /5-HT2 antagonism, cariprazine is also a D3 partial agonist with selectivity for the D3 receptor. Cariprazine’s selectivity for the D 3 receptor may be associated with greater effects on the negative symptoms of schizophrenia. This drug is currently under review for possible approval in 2014. F. Glutamatergic Antipsychotics No glutamate-specific agents are currently approved for the treatment of schizophrenia. However, several agents are in late clinical testing. Among these is bitopertin, a glycine transporter 1 receptor inhibitor (GlyT1). Glycine is a required co-agonist with glutamate at NMDA receptors. Phase 2 studies indicated that bitopertin used adjunctively with standard antipsychotics significantly improved negative symptoms of schizophrenia. Sarcoserine (N-methylglycine), another GlyT1 inhibitor, in combination with a standard antipsychotic has also shown benefit in improving both negative and positive symptoms of schizophrenia in acutely ill as well as in more chronic patients with schizophrenia. Another class of investigational antipsychotic agents includes the metabotropic glutamate receptor agonists. Eight metabotropic glutamate receptors are divided into three groups: group I (mGluR1,5), group II (mGluR2,3), and group III (mGluR4,6,7,8). mGluR2,3 inhibits glutamate release presynaptically. Several mGluR2,3 agents are being investigated in the treatment of schizophrenia. One agent, pomaglumetad methionil, showed antipsychotic efficacy in early phase 2 trials, but subsequent trials failed to show benefit in either positive or negative symptoms of schizophrenia. Other metabotropic glutamate receptor agonists are being explored for the treatment of negative and cognitive symptoms of schizophrenia.

Pharmacokinetics A. Absorption and Distribution Most antipsychotic drugs are readily but incompletely absorbed. Furthermore, many undergo significant first-pass metabolism. Thus, oral doses of chlorpromazine and thioridazine have systemic availability of 25–35%, whereas haloperidol, which has less first-pass metabolism, has an average systemic availability of about 65%. Most antipsychotic drugs are highly lipid soluble and protein bound (92–99%). They tend to have large volumes of distribution (usually more than 7 L/kg). They generally have a much longer clinical duration of action than would be estimated from their plasma half-lives. This is paralleled by prolonged occupancy of D2 dopamine receptors in the brain by the typical antipsychotic drugs. Metabolites of chlorpromazine may be excreted in the urine weeks after the last dose of chronically administered drug. Long-acting injectable formulations may cause some blockade of D2 receptors 3–6 months after the last injection. Time to recurrence of psychotic symptoms is highly variable after discontinuation of antipsychotic drugs. The average time for relapse in stable patients with schizophrenia who discontinue their medication is 6 months. Clozapine is an exception in that relapse after discontinuation is usually rapid and severe. Thus, clozapine should never be discontinued abruptly unless clinically needed because of adverse effects such as


myocarditis or agranulocytosis, which are true medical emergencies. B. Metabolism Most antipsychotic drugs are almost completely metabolized by oxidation or demethylation, catalyzed by liver microsomal cytochrome P450 enzymes. CYP2D6, CYP1A2, and CYP3A4 are the major isoforms involved (see Chapter 4). Drug-drug interactions should be considered when combining antipsychotic drugs with various other psychotropic drugs or drugs—such as ketoconazole—that inhibit various cytochrome P450 enzymes. At the typical clinical doses, antipsychotic drugs do not usually interfere with the metabolism of other drugs.

Pharmacodynamics The first phenothiazine antipsychotic drugs, with chlorpromazine as the prototype, proved to have a wide variety of central nervous system, autonomic, and endocrine effects. Although efficacy of these drugs is primarily driven by D 2 -receptor blockade, their adverse actions were traced to blocking effects at a wide range of receptors including α adrenoceptors and muscarinic, H1 histaminic, and 5-HT2 receptors. A. Dopaminergic Systems Five dopaminergic systems or pathways are important for understanding schizophrenia and the mechanism of action of antipsychotic drugs. The first pathway—the one most closely related to behavior and psychosis—is the mesolimbic-mesocortical pathway, which projects from cell bodies in the ventral tegmentum in separate bundles of axons to the limbic system and neocortex. The second system —the nigrostriatal pathway—consists of neurons that project from the substantia nigra to the dorsal striatum, which includes the caudate and putamen; it is involved in the coordination of voluntary movement. Blockade of the D2 receptors in the nigrostriatal pathway is responsible for EPS. The third pathway—the tuberoinfundibular system—arises in the arcuate nuclei and periventricular neurons and releases dopamine into the pituitary portal circulation. Dopamine released by these neurons physiologically inhibits prolactin secretion from the anterior pituitary. The fourth dopaminergic system—the medullary-periventricular pathway—consists of neurons in the motor nucleus of the vagus whose projections are not well defined. This system may be involved in eating behavior. The fifth pathway— the incertohypothalamic pathway—forms connections from the medial zona incerta to the hypothalamus and the amygdala. It appears to regulate the anticipatory motivational phase of copulatory behavior in rats. After dopamine was identified as a neurotransmitter in 1959, it was shown that its effects on electrical activity in central synapses and on production of the second messenger cAMP synthesized by adenylyl cyclase could be blocked by antipsychotic drugs such as chlorpromazine, haloperidol, and thiothixene. This evidence led to the conclusion in the early 1960s that these drugs should be considered dopamine-receptor antagonists and was a key factor in the development of the dopamine hypothesis of schizophrenia described earlier in this chapter. The antipsychotic action is now thought to be produced (at least in part) by their ability to block the effect of dopamine to inhibit the activity of adenylyl cyclase in the mesolimbic system. B. Dopamine Receptors and Their Effects At present, five dopamine receptors have been described, consisting of two separate families, the D1 -like and D2 -like receptor groups. The D1 receptor is coded by a gene on chromosome 5, increases cAMP by Gs-coupled activation of adenylyl cyclase, and is located mainly in the putamen, nucleus accumbens, and olfactory tubercle and cortex. The other member of this family, D 5 , is coded by a gene on chromosome 4, also increases cAMP, and is found in the hippocampus and hypothalamus. The therapeutic potency of antipsychotic drugs does not correlate with their affinity for binding to the D1 receptor (Figure 29–3, top) nor did a selective D1 antagonist prove to be an effective antipsychotic in patients with schizophrenia. The D2 receptor is coded on chromosome 11, decreases cAMP (by G i-coupled inhibition of adenylyl cyclase), and inhibits calcium channels but opens potassium channels. It is found both pre- and postsynaptically on neurons in the caudate-putamen, nucleus accumbens, and olfactory tubercle. A second member of this family, the D 3 receptor, also coded by a gene on chromosome 11, is thought to also decrease cAMP and is located in the frontal cortex, medulla, and midbrain. D4 receptors also decrease cAMP and are concentrated in the cortex.


FIGURE 29–3 Correlations between the therapeutic potency of antipsychotic drugs and their affinity for binding to dopamine D1 (top) or D2 receptors (bottom). Potency is indicated on the horizontal axes; it decreases to the right. Binding affinity for D1 receptors was measured by displacing the selective D1 ligand SCH 23390; affinity for D2 receptors was similarly measured by displacing the selective D2 ligand haloperidol. Binding affinity decreases upward. (Reprinted, with permission, of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc., from Seeman P: Dopamine receptors and the dopamine hypothesis of schizophrenia. Synapse 1987;1:133.) The typical antipsychotic agents block D2 receptors stereoselectively for the most part, and their binding affinity is very strongly


correlated with clinical antipsychotic and extrapyramidal potency (Figure 29–3, bottom). In vivo imaging studies of D2 -receptor occupancy indicate that for antipsychotic efficacy, the typical antipsychotic drugs must be given in sufficient doses to achieve at least 60% occupancy of striatal D2 receptors. This is not required for the atypical antipsychotic drugs such as clozapine and olanzapine, which are effective at lower occupancy levels of 30–50%, most likely because of their concurrent high occupancy of 5-HT2A receptors. The typical antipsychotic drugs produce EPS when the occupancy of striatal D2 receptors reaches 80% or higher. Positron emission tomography (PET) studies with aripiprazole show very high occupancy of D2 receptors, but this drug does not cause EPS because it is a partial D2 -receptor agonist. Aripiprazole also gains therapeutic efficacy through its 5-HT 2A antagonism and possibly 5-HT1A partial agonism. These findings have been incorporated into the dopamine hypothesis of schizophrenia. However, additional factors complicate interpretation of dopamine receptor data. For example, dopamine receptors exist in both high- and low-affinity forms, and it is not known whether schizophrenia or the antipsychotic drugs alter the proportions of receptors in these two forms. It has not been convincingly demonstrated that antagonism of any dopamine receptor other than the D2 receptor plays a role in the action of antipsychotic drugs. Selective and relatively specific D1 -, D3 -, and D4 -receptor antagonists have been tested repeatedly with no evidence of antipsychotic action. Most of the newer atypical antipsychotic agents and some of the traditional ones have a higher affinity for the 5-HT2A receptor than for the D2 receptor (Table 29–1), suggesting an important role for the serotonin 5-HT system in the etiology of schizophrenia and the action of these drugs. C. Differences among Antipsychotic Drugs Although all effective antipsychotic drugs block D2 receptors, the degree of this blockade in relation to other actions on receptors varies considerably among drugs. Vast numbers of ligand-receptor binding experiments have been performed in an effort to discover a single receptor action that would best predict antipsychotic efficacy. A summary of the relative receptor-binding affinities of several key agents in such comparisons illustrates the difficulty in drawing simple conclusions from such experiments: Chlorpromazine: α1 = 5-HT2A > D2 > D1 Haloperidol: D2 > α1 > D4 > 5-HT2A > D1 > H1 Clozapine: D4 = α1 > 5-HT2A > D2 = D1 Olanzapine: 5-HT2A > H1 > D4 > D2 > α1 > D1 Aripiprazole: D2 = 5-HT2A > D4 > α1 = H1 >> D1 Quetiapine: H1 > α1 > M1,3 > D2 > 5-HT2A Thus, most of the atypical and some typical antipsychotic agents are at least as potent in inhibiting 5-HT2 receptors as they are in inhibiting D2 receptors. The newest, aripiprazole, appears to be a partial agonist of D2 receptors. Varying degrees of antagonism of α 2 adrenoceptors are also seen with risperidone, clozapine, olanzapine, quetiapine, and aripiprazole. Current research is directed toward discovering atypical antipsychotic compounds that are either more selective for the mesolimbic system (to reduce their effects on the extrapyramidal system) or have effects on central neurotransmitter receptors—such as those for acetylcholine and excitatory amino acids—that have been proposed as new targets for antipsychotic action. In contrast to the difficult search for receptors responsible for antipsychotic efficacy, the differences in receptor effects of various antipsychotics do explain many of their toxicities (Tables 29–1 and 29–2). In particular, extrapyramidal toxicity appears to be consistently associated with high D2 potency. TABLE 29–2 Adverse pharmacologic effects of antipsychotic drugs.


D. Psychological Effects Most antipsychotic drugs cause unpleasant subjective effects in nonpsychotic individuals. The mild to severe EPS, including akathisia, sleepiness, restlessness, and autonomic effects are unlike any associated with more familiar sedatives or hypnotics. Nevertheless, low doses of some of these drugs, particularly quetiapine, are used to promote sleep onset and maintenance, although there is no approved indication for such usage. People without psychiatric illness given antipsychotic drugs, even at low doses, experience impaired performance as judged by a number of psychomotor and psychometric tests. Psychotic individuals, however, may actually show improvement in their performance as the psychosis is alleviated. The ability of the atypical antipsychotic drugs to improve some domains of cognition in patients with schizophrenia and bipolar disorder is controversial. Some individuals experience marked improvement, and for that reason, cognition should be assessed in all patients with schizophrenia and a trial of an atypical agent considered, even if positive symptoms are well controlled by typical agents. E. Electroencephalographic Effects Antipsychotic drugs produce shifts in the pattern of electroencephalographic (EEG) frequencies, usually slowing them and increasing their synchronization. The slowing (hypersynchrony) is sometimes focal or unilateral, which may lead to erroneous diagnostic interpretations. Both the frequency and the amplitude changes induced by psychotropic drugs are readily apparent and can be quantitated


by sophisticated electrophysiologic techniques. Some of the neuroleptic agents lower the seizure threshold and induce EEG patterns typical of seizure disorders; however, with careful dosage titration, most can be used safely in epileptic patients. F. Endocrine Effects Older typical antipsychotic drugs, as well as risperidone and paliperidone, produce elevations of prolactin (see Adverse Effects, below). Newer antipsychotics such as olanzapine, quetiapine, and aripiprazole cause no or minimal increases of prolactin and reduced risks of extrapyramidal system dysfunction and tardive dyskinesia, reflecting their diminished D2 antagonism. G. Cardiovascular Effects The low-potency phenothiazines frequently cause orthostatic hypotension and tachycardia. Mean arterial pressure, peripheral resistance, and stroke volume are decreased. These effects are predictable from the autonomic actions of these agents (Table 29–2). Abnormal electrocardiograms have been recorded, especially with thioridazine. Changes include prolongation of QT interval and abnormal configurations of the ST segment and T waves. These changes are readily reversed by withdrawing the drug. Thioridazine, however, is not associated with increased risk of torsades more than other typical antipsychotics, whereas haloperidol, which does not increase QTc, is. Among the newest atypical antipsychotics, prolongation of the QT or QTc interval has received much attention. Because this was believed to indicate an increased risk of dangerous arrhythmias, approval of sertindole has been delayed and ziprasidone and quetiapine are accompanied by warnings. There is, however, no evidence that this has actually translated into increased incidence of arrhythmias. The atypical antipsychotics are also associated with a metabolic syndrome that may increase the risk of coronary artery disease, stroke, and hypertension.

CLINICAL PHARMACOLOGY OF ANTIPSYCHOTIC AGENTS Indications A. Psychiatric Indications Schizophrenia is the primary indication for antipsychotic agents. However, in the last decade, the use of antipsychotics in the treatment of mood disorders such as bipolar disorder (BP1), psychotic depression, and treatment-resistant depression has eclipsed their use in the treatment of schizophrenia. Catatonic forms of schizophrenia are best managed by intravenous benzodiazepines. Antipsychotic drugs may be needed to treat psychotic components of that form of the illness after catatonia has ended, and they remain the mainstay of treatment for this condition. Unfortunately, many patients show little response, and virtually none show a complete response. Antipsychotic drugs are also indicated for schizoaffective disorders, which share characteristics of both schizophrenia and affective disorders. No fundamental difference between these two diagnoses has been reliably demonstrated. It is most likely that they are part of a continuum with bipolar psychotic disorder. The psychotic aspects of the illness require treatment with antipsychotic drugs, which may be used with other drugs such as antidepressants, lithium, or valproic acid. The manic phase in bipolar affective disorder often requires treatment with antipsychotic agents, although lithium or valproic acid supplemented with high-potency benzodiazepines (eg, lorazepam or clonazepam) may suffice in milder cases. Recent controlled trials support the efficacy of monotherapy with atypical antipsychotics in the acute phase (up to 4 weeks) of mania. In addition, several second generation antipsychotics are approved in the maintenance treatment of bipolar disorder. They appear more effective in preventing mania than in preventing depression. As mania subsides, the antipsychotic drug may be withdrawn, although maintenance treatment with atypical antipsychotic agents has become more common. Nonmanic excited states may also be managed by antipsychotics, often in combination with benzodiazepines. An increasingly common use of antipsychotics is in the monotherapy of acute bipolar depression and the adjunctive use of antipsychotics with antidepressants in the treatment of unipolar depression. Several antipsychotics are now FDA approved in the management of bipolar depression including quetiapine, lurasidone, and olanzapine (in a combination formulation with fluoxetine). The antipsychotics appear more consistently effective than antidepressants in the treatment of bipolar depression and also do not increase the risk of inducing mania or increasing the frequency of bipolar cycling. Likewise, several antipsychotics, including aripiprazole, quetiapine, and olanzapine, are now approved in the adjunctive treatment of unipolar depression. Although many drugs are combined with antidepressants in the adjunctive treatment of major depression, antipsychotic agents are the only class of agents that have been formally evaluated for possible approval for this purpose. Residual symptoms and partial remission are common, with antidepressants showing consistent benefit in improving overall antidepressant response. Some of the intramuscular antipsychotics have been approved for the control of agitation associated with bipolar disorder and schizophrenia. Antipsychotics such as haloperidol have long been used in the ICU setting to manage agitation in delirious and postsurgical patients. The intramuscular forms of ziprasidone and aripiprazole have been shown to improve agitation within 1–2 hours, with fewer extrapyramidal symptoms than typical agents such as haloperidol.


Other indications for the use of antipsychotics include Tourette’s syndrome and possibly disturbed behavior in patients with Alzheimer’s disease. However, controlled trials of antipsychotics in the management of behavioral symptoms in dementia patients have generally not demonstrated efficacy. Furthermore, second-generation as well as some first-generation antipsychotics have been associated with increased mortality in these patients. Antipsychotics are not indicated for the treatment of various withdrawal syndromes, eg, opioid withdrawal. In small doses, antipsychotic drugs have been promoted (wrongly) for the relief of anxiety associated with minor emotional disorders. The antianxiety sedatives (see Chapter 22) are preferred in terms of both safety and acceptability to patients. B. Nonpsychiatric Indications Most older typical antipsychotic drugs, with the exception of thioridazine, have a strong antiemetic effect. This action is due to dopamine-receptor blockade, both centrally (in the chemoreceptor trigger zone of the medulla) and peripherally (on receptors in the stomach). Some drugs, such as prochlorperazine and benzquinamide, are promoted solely as antiemetics. Phenothiazines with shorter side chains have considerable H1 -receptor-blocking action and have been used for relief of pruritus or, in the case of promethazine, as preoperative sedatives. The butyrophenone droperidol is used in combination with an opioid, fentanyl, in neuroleptanesthesia. The use of these drugs in anesthesia practice is described in Chapter 25.

Drug Choice Choice among antipsychotic drugs is based mainly on differences in adverse effects and possible differences in efficacy. In addition, cost and the availability of a given agent on drug formularies also influence the choice of a specific antipsychotic. Because use of the older drugs is still widespread, especially for patients treated in the public sector, knowledge of such agents as chlorpromazine and haloperidol remains relevant. Thus, one should be familiar with one member of each of the three subfamilies of phenothiazines, a member of the thioxanthene and butyrophenone group, and all of the newer compounds—clozapine, risperidone, olanzapine, quetiapine, ziprasidone, and aripiprazole. Each may have special advantages for selected patients. A representative group of antipsychotic drugs is presented in Table 29–3. TABLE 29–3 Some representative antipsychotic drugs.



For approximately 70% of patients with schizophrenia, and probably for a similar proportion of patients with bipolar disorder with psychotic features, typical and atypical antipsychotic drugs are of equal efficacy for treating positive symptoms. However, the evidence favors atypical drugs for benefit for negative symptoms and cognition, for diminished risk of tardive dyskinesia and other forms of EPS, and for lesser increases in prolactin levels. Some of the atypical antipsychotic drugs produce more weight gain and increases in lipids than some typical antipsychotic drugs. A small percentage of patients develop diabetes mellitus, most often seen with clozapine and olanzapine. Ziprasidone is the atypical drug causing the least weight gain. Risperidone, paliperidone, and aripiprazole usually produce small increases in weight and lipids. Asenapine and quetiapine have an intermediate effect. Clozapine and olanzapine frequently result in large increases in weight and lipids. Thus, these drugs should be considered as second-line drugs unless there is a specific indication. That is the case with clozapine, which at high doses (300–900 mg/d) is effective in the majority of patients with schizophrenia refractory to other drugs, provided that treatment is continued for up to 6 months. Case reports and several clinical trials suggest that high-dose olanzapine, ie, doses of 30–45 mg/d, may also be efficacious in refractory schizophrenia when given over a 6-month period. Clozapine is the only atypical antipsychotic drug indicated to reduce the risk of suicide. All patients with schizophrenia who have made life-threatening suicide attempts should be seriously evaluated for switching to clozapine. New antipsychotic drugs have been shown in some trials to be more effective than older ones for treating negative symptoms. The floridly psychotic form of the illness accompanied by uncontrollable behavior probably responds equally well to all potent antipsychotics but is still frequently treated with older drugs that offer intramuscular formulations for acute and chronic treatment. Moreover, the low cost of the older drugs contributes to their widespread use despite their risk of adverse EPS effects. Several of the newer antipsychotics, including clozapine, risperidone, and olanzapine, show superiority over haloperidol in terms of overall response in some controlled trials. More comparative studies with aripiprazole are needed to evaluate its relative efficacy. Moreover, the superior adverse-effect profile of the newer agents and low to absent risk of tardive dyskinesia suggest that these should provide the first line of treatment. The best guide for selecting a drug for an individual patient is the patient’s past responses to drugs. At present, clozapine is limited to those patients who have failed to respond to substantial doses of conventional antipsychotic drugs. The agranulocytosis and seizures associated with this drug prevent more widespread use. Risperidone’s superior adverse-effect profile (compared with that of haloperidol) at dosages of 6 mg/d or less and the lower risk of tardive dyskinesia have contributed to its widespread use. Olanzapine and quetiapine may have even lower risk and have also achieved widespread use.

Dosage The range of effective dosages among various antipsychotic agents is broad. Therapeutic margins are substantial. At appropriate dosages, antipsychotics—with the exception of clozapine and perhaps olanzapine—are of equal efficacy in broadly selected groups of patients. However, some patients who fail to respond to one drug may respond to another; for this reason, several drugs may have to be tried to find the one most effective for an individual patient. Patients who have become refractory to two or three antipsychotic agents given in substantial doses become candidates for treatment with clozapine or high-dose olanzapine. Thirty to fifty percent of patients previously refractory to standard doses of other antipsychotic drugs respond to these drugs. In such cases, the increased risk of clozapine can well be justified. Some dosage relationships between various antipsychotic drugs, as well as possible therapeutic ranges, are shown in Table 29–4. TABLE 29–4 Dose relationships of antipsychotics.


Parenteral Preparations Well-tolerated parenteral forms of the high-potency older drugs haloperidol and fluphenazine are available for rapid initiation of treatment as well as for maintenance treatment in noncompliant patients. Since the parenterally administered drugs may have much greater bioavailability than the oral forms, doses should be only a fraction of what might be given orally, and the manufacturer’s literature should be consulted. Fluphenazine decanoate and haloperidol decanoate are suitable for long-term parenteral maintenance therapy in patients who cannot or will not take oral medication. In addition, newer long-acting injectable (LAI) second-generation antipsychotics are now available, including formulations of risperidone, olanzapine, aripiprazole, and paliperidone. For some patients, the newer LAI drugs may be better tolerated than the older depot injectables.

Dosage Schedules Antipsychotic drugs are often given in divided daily doses, titrating to an effective dosage. The low end of the dosage range in Table 29– 4 should be tried for at least several weeks. After an effective daily dosage has been defined for an individual patient, doses can be given


less frequently. Once-daily doses, usually given at night, are feasible for many patients during chronic maintenance treatment. Simplification of dosage schedules leads to better compliance.

Maintenance Treatment A very small minority of schizophrenic patients may recover from an acute episode and require no further drug therapy for prolonged periods. In most cases, the choice is between “as needed” increased doses or the addition of other drugs for exacerbations versus continual maintenance treatment with full therapeutic dosage. The choice depends on social factors such as the availability of family or friends familiar with the early symptoms of relapse and ready access to care.

Drug Combinations Combining antipsychotic drugs confounds evaluation of the efficacy of the drugs being used. Use of combinations, however, is widespread, with more emerging experimental data supporting such practices. Tricyclic antidepressants or, more often, selective serotonin reuptake inhibitors (SSRIs) are often used with antipsychotic agents for symptoms of depression complicating schizophrenia. The evidence for the usefulness of this polypharmacy is minimal. Electroconvulsive therapy (ECT) is a useful adjunct for antipsychotic drugs, not only for treating mood symptoms, but for positive symptom control as well. Electroconvulsive therapy can augment clozapine when maximum doses of clozapine are ineffective. In contrast, adding risperidone to clozapine is not beneficial. Lithium or valproic acid is sometimes added to antipsychotic agents with benefit to patients who do not respond to the latter drugs alone. There is some evidence that lamotrigine is more effective than any of the other mood stabilizers for this indication (see below). It is uncertain whether instances of successful combination therapy represent misdiagnosed cases of mania or schizoaffective disorder. Benzodiazepines may be useful for patients with anxiety symptoms or insomnia not controlled by antipsychotics.

Adverse Reactions Most of the unwanted effects of antipsychotic drugs are extensions of their known pharmacologic actions (Tables 29–1 and 29–2), but a few effects are allergic in nature and some are idiosyncratic. A. Behavioral Effects The older typical antipsychotic drugs are unpleasant to take. Many patients stop taking these drugs because of the adverse effects, which may be mitigated by giving small doses during the day and the major portion at bedtime. A “pseudodepression” that may be due to druginduced akinesia usually responds to cautious treatment with antiparkinsonism drugs. Other pseudodepressions may be due to higher doses than needed in a partially remitted patient, in which case decreasing the dose may relieve the symptoms. Toxic-confusional states may occur with very high doses of drugs that have prominent antimuscarinic actions. B. Neurologic Effects Extrapyramidal reactions occurring early during treatment with older agents include typical Parkinson’s syndrome, akathisia (uncontrollable restlessness), and acute dystonic reactions (spastic retrocollis or torticollis). Parkinsonism can be treated, when necessary, with conventional antiparkinsonism drugs of the antimuscarinic type or, in rare cases, with amantadine. (Levodopa should never be used in these patients.) Parkinsonism may be self-limiting, so that an attempt to withdraw antiparkinsonism drugs should be made every 3–4 months. Akathisia and dystonic reactions also respond to such treatment, but many clinicians prefer to use a sedative antihistamine with anticholinergic properties, eg, diphenhydramine, which can be given either parenterally or orally. Tardive dyskinesia, as the name implies, is a late-occurring syndrome of abnormal choreoathetoid movements. It is the most important unwanted effect of antipsychotic drugs. It has been proposed that it is caused by a relative cholinergic deficiency secondary to supersensitivity of dopamine receptors in the caudate-putamen. The prevalence varies enormously, but tardive dyskinesia is estimated to have occurred in 20–40% of chronically treated patients before the introduction of the newer atypical antipsychotics. Early recognition is important, since advanced cases may be difficult to reverse. Any patient with tardive dyskinesia treated with a typical antipsychotic drug or possibly risperidone or paliperidone should be switched to quetiapine or clozapine, the atypical agents with the least likelihood of causing tardive dyskinesia. Many treatments have been proposed, but their evaluation is confounded by the fact that the course of the disorder is variable and sometimes self-limited. Reduction in dosage may also be considered. Most authorities agree that the first step should be to discontinue or reduce the dose of the current antipsychotic agent or switch to one of the newer atypical agents. A logical second step would be to eliminate all drugs with central anticholinergic action, particularly antiparkinsonism drugs and tricyclic antidepressants. These two steps are often enough to bring about improvement. If they fail, the addition of diazepam in doses as high as 30–40 mg/d may add to the improvement by enhancing GABAergic activity. Seizures, though recognized as a complication of chlorpromazine treatment, were so rare with the high-potency older drugs as to merit little consideration. However, de novo seizures may occur in 2–5% of patients treated with clozapine. Use of an anticonvulsant is


able to control seizures in most cases. C. Autonomic Nervous System Effects Most patients are able to tolerate the antimuscarinic adverse effects of antipsychotic drugs. Those who are made too uncomfortable or who develop urinary retention or other severe symptoms can be switched to an agent without significant antimuscarinic action. Orthostatic hypotension or impaired ejaculation—common complications of therapy with chlorpromazine or mesoridazine—should be managed by switching to drugs with less marked adrenoceptor-blocking actions. D. Metabolic and Endocrine Effects Weight gain is very common, especially with clozapine and olanzapine, and requires monitoring of food intake, especially carbohydrates. Hyperglycemia may develop, but whether secondary to weight gain-associated insulin resistance or to other potential mechanisms remains to be clarified. Hyperlipidemia may occur. The management of weight gain, insulin resistance, and increased lipids should include monitoring of weight at each visit and measurement of fasting blood sugar and lipids at 3- to 6-month intervals. Measurement of hemoglobin A 1C may be useful when it is impossible to be sure of obtaining a fasting blood sugar. Diabetic ketoacidosis has been reported in a few cases. The triglyceride:HDL ratio should be less than 3.5 in fasting samples. Levels higher than that indicate increased risk of atherosclerotic cardiovascular disease. Hyperprolactinemia in women results in the amenorrhea-galactorrhea syndrome and infertility; in men, loss of libido, impotence, and infertility may result. Hyperprolactinemia may cause osteoporosis, particularly in women. If dose reduction is not indicated, or ineffective in controlling this pattern, switching to one of the atypical agents that do not raise prolactin levels, eg, aripiprazole, may be indicated. E. Toxic or Allergic Reactions Agranulocytosis, cholestatic jaundice, and skin eruptions occur rarely with the high-potency antipsychotic drugs currently used. In contrast to other antipsychotic agents, clozapine causes agranulocytosis in a small but significant number of patients— approximately 1–2% of those treated. This serious, potentially fatal effect can develop rapidly, usually between the 6th and 18th weeks of therapy. It is not known whether it represents an immune reaction, but it appears to be reversible upon discontinuance of the drug. Because of the risk of agranulocytosis, patients receiving clozapine must have weekly blood counts for the first 6 months of treatment and every 3 weeks thereafter. F. Ocular Complications Deposits in the anterior portions of the eye (cornea and lens) are a common complication of chlorpromazine therapy. They may accentuate the normal processes of aging of the lens. Thioridazine is the only antipsychotic drug that causes retinal deposits, which in advanced cases may resemble retinitis pigmentosa. The deposits are usually associated with “browning” of vision. The maximum daily dose of thioridazine has been limited to 800 mg/d to reduce the possibility of this complication. G. Cardiac Toxicity Thioridazine in doses exceeding 300 mg daily is almost always associated with minor abnormalities of T waves that are easily reversible. Overdoses of thioridazine are associated with major ventricular arrhythmias, eg, torsades de pointes, cardiac conduction block, and sudden death; it is not certain whether thioridazine can cause these same disorders when used in therapeutic doses. In view of possible additive antimuscarinic and quinidine-like actions with various tricyclic antidepressants, thioridazine should be combined with the latter drugs only with great care. Among the atypical agents, ziprasidone carries the greatest risk of QT prolongation and therefore should not be combined with other drugs that prolong the QT interval, including thioridazine, pimozide, and group 1A or 3 antiarrhythmic drugs. Clozapine is sometimes associated with myocarditis and must be discontinued if myocarditis manifests. Sudden death due to arrhythmias is common in schizophrenia. It is not always drug-related, and there are no studies that definitively show increased risk with particular drugs. Monitoring of QTc prolongation has proved to be of little use unless the values increase to more than 500 msec and this is manifested in multiple rhythm strips or a Holter monitor study. A 20,000-patient study of ziprasidone versus olanzapine showed minimal or no increased risk of torsades de pointes or sudden death in patients who were randomized to ziprasidone. H. Use in Pregnancy; Dysmorphogenesis Although antipsychotic drugs appear to be relatively safe in pregnancy, a small increase in teratogenic risk could be missed. Questions about whether to use these drugs during pregnancy and whether to abort a pregnancy in which the fetus has already been exposed must be decided individually. If a pregnant woman could manage to be free of antipsychotic drugs during pregnancy, this would be desirable because of their effects on the neurotransmitters involved in neurodevelopment. I. Neuroleptic Malignant Syndrome This life-threatening disorder occurs in patients who are extremely sensitive to the extrapyramidal effects of antipsychotic agents (see


a ls o Chapter 16). The initial symptom is marked muscle rigidity. If sweating is impaired, as it often is during treatment with anticholinergic drugs, fever may ensue, often reaching dangerous levels. The stress leukocytosis and high fever associated with this syndrome may erroneously suggest an infectious process. Autonomic instability, with altered blood pressure and pulse rate, is often present. Muscle-type creatine kinase levels are usually elevated, reflecting muscle damage. This syndrome is believed to result from an excessively rapid blockade of postsynaptic dopamine receptors. A severe form of extrapyramidal syndrome follows. Early in the course, vigorous treatment of the extrapyramidal syndrome with antiparkinsonism drugs is worthwhile. Muscle relaxants, particularly diazepam, are often useful. Other muscle relaxants, such as dantrolene, or dopamine agonists, such as bromocriptine, have been reported to be helpful. If fever is present, cooling by physical measures should be tried. Various minor forms of this syndrome are now recognized. Switching to an atypical drug after recovery is indicated.

Drug Interactions Antipsychotics produce more important pharmacodynamic than pharmacokinetic interactions because of their multiple effects. Additive effects may occur when these drugs are combined with others that have sedative effects, α-adrenoceptor-blocking action, anticholinergic effects, and—for thioridazine and ziprasidone—quinidine-like action. A variety of pharmacokinetic interactions have been reported, but none are of major clinical significance.

Overdoses Poisonings with antipsychotic agents (unlike tricyclic antidepressants) are rarely fatal, with the exception of those due to mesoridazine and thioridazine. In general, drowsiness proceeds to coma, with an intervening period of agitation. Neuromuscular excitability may be increased and proceed to convulsions. Pupils are miotic, and deep tendon reflexes are decreased. Hypotension and hypothermia are the rule, although fever may be present later in the course. The lethal effects of mesoridazine and thioridazine are related to induction of ventricular tachyarrhythmias. Patients should be given the usual “ABCD” treatment for poisonings (see Chapter 58) and treated supportively. Management of overdoses of thioridazine and mesoridazine, which are complicated by cardiac arrhythmias, is similar to that for tricyclic antidepressants (see Chapter 30).

Psychosocial Treatment & Cognitive Remediation Patients with schizophrenia need psychosocial support based around activities of daily living, including housing, social activities, returning to school, obtaining the optimal level of work they may be capable of, and restoring social interactions. Unfortunately, funding for this crucial component of treatment has been minimized in recent years. Case management and therapy services are a vital part of the treatment program that should be provided to patients with schizophrenia. First-episode patients are particularly needful of this support because they often deny their illness and are noncompliant with medication.

Benefits & Limitations of Drug Treatment As noted at the beginning of this chapter, antipsychotic drugs have had a major impact on psychiatric treatment. First, they have shifted the vast majority of patients from long-term hospitalization to the community. For many patients, this shift has provided a better life under more humane circumstances and in many cases has made possible life without frequent use of physical restraints. For others, the tragedy of an aimless existence is now being played out in the streets of our communities rather than in mental institutions. Second, these antipsychotic drugs have markedly shifted psychiatric thinking to a more biologic orientation. Partly because of research stimulated by the effects of these drugs on schizophrenia, we now know much more about central nervous system physiology and pharmacology than was known before the introduction of these agents. However, despite much research, schizophrenia remains a scientific mystery and a personal disaster for the patient. Although most schizophrenic patients obtain some degree of benefit from these drugs—in some cases substantial benefit—none are made well by them.

LITHIUM, MOOD-STABILIZING DRUGS, & OTHER TREATMENT FOR BIPOLAR DISORDER Bipolar disorder, once known as manic-depressive illness, was conceived of as a psychotic disorder distinct from schizophrenia at the end of the 19th century. Before that both of these disorders were considered part of a continuum. It is ironic that the weight of the evidence today is that there is profound overlap in these disorders. This is not to say that there are no pathophysiologically important differences or that some drug treatments are differentially effective in these disorders. According to DSM-IV, they are separate disease entities while research continues to define the dimensions of these illnesses and their genetic and other biologic markers.


Lithium was the first agent shown to be useful in the treatment of the manic phase of bipolar disorder that was not also an antipsychotic drug. Lithium has no known use in schizophrenia. Lithium continues to be used for acute-phase illness as well as for prevention of recurrent manic and depressive episodes. A group of mood-stabilizing drugs that are also anticonvulsant agents has become more widely used than lithium. It includes carbamazepine and valproic acid for the treatment of acute mania and for prevention of its recurrence. Lamotrigine is approved for prevention of recurrence. Gabapentin, oxcarbazepine, and topiramate are sometimes used to treat bipolar disorder but are not approved by the FDA for this indication. Aripiprazole, chlorpromazine, olanzapine, quetiapine, risperidone, and ziprasidone are approved by the FDA for treatment of the manic phase of bipolar disorder. Olanzapine plus fluoxetine in combination and quetiapine are approved for treatment of bipolar depression.

Nature of Bipolar Affective Disorder Bipolar affective disorder occurs in 1–3% of the adult population. It may begin in childhood, but most cases are first diagnosed in the third and fourth decades of life. The key symptoms of bipolar disorder in the manic phase are expansive or irritable mood, hyperactivity, impulsivity, disinhibition, diminished need for sleep, racing thoughts, psychotic symptoms in some (but not all) patients, and cognitive impairment. Depression in bipolar patients is phenomenologically similar to that of major depression, with the key features being depressed mood, diurnal variation, sleep disturbance, anxiety, and sometimes, psychotic symptoms. Mixed manic and depressive symptoms are also seen. Patients with bipolar disorder are at high risk for suicide. The sequence, number, and intensity of manic and depressive episodes are highly variable. The cause of the mood swings characteristic of bipolar affective disorder is unknown, although a preponderance of catecholamine-related activity may be present. Drugs that increase this activity tend to exacerbate mania, whereas those that reduce activity of dopamine or norepinephrine relieve mania. Acetylcholine or glutamate may also be involved. The nature of the abrupt switch from mania to depression experienced by some patients is uncertain. Bipolar disorder has a strong familial component, and there is abundant evidence that bipolar disorder is genetically determined. Many of the genes that increase vulnerability to bipolar disorder are common to schizophrenia but some genes appear to be unique to each disorder. Genome-wide association studies of psychotic bipolar disorder have shown replicated linkage to chromosomes 8p and 13q. Several candidate genes have shown association with bipolar disorder with psychotic features and with schizophrenia. These include genes for dysbindin, DAOA/G30, disrupted-in-schizophrenia-1 (DISC-1), and neuregulin 1.

BASIC PHARMACOLOGY OF LITHIUM Lithium was first used therapeutically in the mid-19th century in patients with gout. It was briefly used as a substitute for sodium chloride in hypertensive patients in the 1940s but was banned after it proved too toxic for use without monitoring. In 1949, Cade discovered that lithium was an effective treatment for bipolar disorder, engendering a series of controlled trials that confirmed its efficacy as monotherapy for the manic phase of bipolar disorder.

Pharmacokinetics Lithium is a small monovalent cation. Its pharmacokinetics are summarized in Table 29–5. TABLE 29–5 Pharmacokinetics of lithium.


Pharmacodynamics Despite considerable investigation, the biochemical basis for mood stabilizer therapies including lithium and anticonvulsant mood stabilizers is not clearly understood. Lithium directly inhibits two signal transduction pathways. It both suppresses inositol signaling through depletion of intracellular inositol and inhibits glycogen synthase kinase-3 (GSK-3), a multifunctional protein kinase. GSK-3 is a component of diverse intracellular signaling pathways. These include signaling via insulin/insulin-like growth factor, brain-derived neurotrophic factor (BDNF), and the Wnt pathway. All of these lead to inhibition of GSK-3. GSK-3 phosphorylates β-catenin, resulting in interaction with transcription factors. The pathways that are facilitated in this manner modulate energy metabolism, provide neuroprotection, and increase neuroplasticity. Studies on the enzyme prolyl oligopeptidase and the sodium myoinositol transporter support an inositol depletion mechanism for moodstabilizer action. Valproic acid may indirectly reduce GSK-3 activity and can up-regulate gene expression through inhibition of histone deacetylase. Valproic acid also inhibits inositol signaling through an inositol depletion mechanism. There is no evidence of GSK-3 inhibition by carbamazepine, a second antiepileptic mood stabilizer. In contrast, this drug alters neuronal morphology through an inositol depletion mechanism, as seen with lithium and valproic acid. The mood stabilizers may also have indirect effects on neurotransmitters and their release. A. Effects on Electrolytes and Ion Transport Lithium is closely related to sodium in its properties. It can substitute for sodium in generating action potentials and in Na+-Na+ exchange across the membrane. It inhibits the latter process; that is, Li+-Na+ exchange is gradually slowed after lithium is introduced into the body. At therapeutic concentrations (~1 mEq/L), it does not significantly affect the Na+-Ca2+ exchanger or the Na+/K+-ATPase pump. B. Effects on Second Messengers Some of the enzymes affected by lithium are listed in Table 29–6. One of the best-defined effects of lithium is its action on inositol phosphates. Early studies of lithium demonstrated changes in brain inositol phosphate levels, but the significance of these changes was not appreciated until the second-messenger roles of inositol-1,4,5-trisphosphate (IP 3 ) and diacylglycerol (DAG) were discovered. As described in Chapter 2, inositol trisphosphate and diacylglycerol are important second messengers for both α-adrenergic and muscarinic transmission. Lithium inhibits inositol monophosphatase (IMPase) and other important enzymes in the normal recycling of membrane phosphoinositides, including conversion of IP 2 (inositol diphosphate) to IP 1 (inositol monophosphate) and the conversion of IP 1 to inositol (Figure 29–4). This block leads to a depletion of free inositol and ultimately of phosphatidylinositol-4,5-bisphosphate (PIP 2 ), the membrane precursor of IP 3 and DAG. Over time, the effects of transmitters on the cell diminish in proportion to the amount of activity in


the PIP 2 -dependent pathways. The activity of these pathways is postulated to be markedly increased during a manic episode. Treatment with lithium would be expected to diminish activity in these circuits. TABLE 29–6 Enzymes affected by lithium at therapeutic concentrations.


FIGURE 29–4 Effect of lithium on the IP 3 (inositol trisphosphate) and DAG (diacylglycerol) second-messenger system. The schematic diagram shows the synaptic membrane of a neuron. (PIP 2 , phosphatidylinositol-4,5-bisphosphate; PLC, phospholipase C; G, coupling protein; Effects, activation of protein kinase C, mobilization of intracellular Ca2+, etc.) Lithium, by inhibiting the recycling of inositol substrates, may cause the depletion of the second-messenger source PIP 2 and therefore reduce the release of IP 3 and DAG. Lithium may also act by other mechanisms. Studies of noradrenergic effects in isolated brain tissue indicate that lithium can inhibit norepinephrine-sensitive adenylyl cyclase. Such an effect could relate to both its antidepressant and its antimanic effects. The relationship of these effects to lithium’s actions on IP 3 mechanisms is currently unknown. Because lithium affects second-messenger systems involving both activation of adenylyl cyclase and phosphoinositol turnover, it is not surprising that G proteins are also found to be affected. Several studies suggest that lithium may uncouple receptors from their G proteins; indeed, two of lithium’s most common side effects, polyuria and subclinical hypothyroidism, may be due to uncoupling of the vasopressin and thyroid-stimulating hormone (TSH) receptors from their G proteins. The major current working hypothesis for lithium’s therapeutic mechanism of action supposes that its effects on phosphoinositol turnover, leading to an early relative reduction of myoinositol in human brain, are part of an initiating cascade of intracellular changes. Effects on specific isoforms of protein kinase C may be most relevant. Alterations of protein kinase C-mediated signaling alter gene expression and the production of proteins implicated in long-term neuroplastic events that could underlie long-term mood stabilization.

CLINICAL PHARMACOLOGY OF LITHIUM Bipolar Affective Disorder Until the late 1990s, lithium carbonate was the universally preferred treatment for bipolar disorder, especially in the manic phase. With the approval of valproate, aripiprazole, olanzapine, quetiapine, risperidone, and ziprasidone for this indication, a smaller percentage of bipolar patients now receive lithium. This trend is reinforced by the slow onset of action of lithium, which has often been supplemented with concurrent use of antipsychotic drugs or potent benzodiazepines in severely manic patients. The overall success rate for achieving remission from the manic phase of bipolar disorder can be as high as 80% but lower among patients who require hospitalization. A similar situation applies to maintenance treatment, which is about 60% effective overall but less in severely ill patients. These considerations have led to increased use of combined treatment in severe cases. After mania is controlled, the antipsychotic drug may be stopped and benzodiazepines and lithium continued as maintenance therapy. The depressive phase of manic-depressive disorder often requires concurrent use of other agents including antipsychotics such as quetiapine or lurasidone. Antidepressants have not shown consistent utility and may be destabilizing. Tricyclic antidepressant agents have been linked to precipitation of mania, with more rapid cycling of mood swings, although most patients do not show this effect. Similarly, SNRI agents (see Chapter 30) have been associated with higher rates of switching to mania than some antidepressants. Selective serotonin reuptake inhibitors are less likely to induce mania but may have limited efficacy. Bupropion has shown some promise but—like tricyclic antidepressants—may induce mania at higher doses. As shown in recent controlled trials, the anticonvulsant lamotrigine is effective for some patients with bipolar depression, but results have been inconsistent. For some patients, however, one of the older monoamine oxidase inhibitors may be the antidepressant of choice. Quetiapine and the combination of olanzapine and fluoxetine has been approved for use in bipolar depression. Unlike antipsychotic or antidepressant drugs, which exert several actions on the central or autonomic nervous system, lithium ion at therapeutic concentrations is devoid of autonomic blocking effects and of activating or sedating effects, although it can produce nausea and tremor. Most important is that the prophylactic use of lithium can prevent both mania and depression. Many experts believe that the


aggressive marketing of newer drugs has inappropriately produced a shift to drugs that are less effective than lithium for substantial numbers of patients.

Other Applications Recurrent depression with a cyclic pattern is controlled by either lithium or imipramine, both of which are superior to placebo. Lithium is also among the better-studied agents used to augment standard antidepressant response in acute major depression in those patients who have had inadequate response to monotherapy. For this application, concentrations of lithium at the lower end of the recommended range for bipolar disorder appear to be adequate. Schizoaffective disorder, another condition with an affective component characterized by a mixture of schizophrenic symptoms and depression or excitement, is treated with antipsychotic drugs alone or combined with lithium. Various antidepressants are added if depression is present. Lithium alone is rarely successful in treating schizophrenia, but adding it to an antipsychotic may salvage an otherwise treatmentresistant patient. Carbamazepine may work equally well when added to an antipsychotic drug.

Monitoring Treatment Clinicians rely on measurements of serum lithium concentrations for assessing both the dosage required for treatment of acute mania and for prophylactic maintenance. These measurements are customarily taken 10–12 hours after the last dose, so all data in the literature pertaining to these concentrations reflect this interval. An initial determination of serum lithium concentration should be obtained about 5 days after the start of treatment, at which time steady-state conditions should have been attained. If the clinical response suggests a change in dosage, simple arithmetic (new dose equals present dose times desired blood level divided by present blood level) should produce the desired level. The serum concentration attained with the adjusted dosage can be checked after another 5 days. Once the desired concentration has been achieved, levels can be measured at increasing intervals unless the schedule is influenced by intercurrent illness or the introduction of a new drug into the treatment program.

Maintenance Treatment The decision to use lithium as prophylactic treatment depends on many factors: the frequency and severity of previous episodes, a crescendo pattern of appearance, and the degree to which the patient is willing to follow a program of indefinite maintenance therapy. Patients with a history of two or more mood cycles or any clearly defined bipolar I diagnosis are probable candidates for maintenance treatment. It has become increasingly evident that each recurrent cycle of bipolar illness may leave residual damage and worsen the long-term prognosis of the patient. Thus, there is greater consensus among experts that maintenance treatment be started as early as possible to reduce the frequency of recurrence. Although some patients can be maintained with serum levels as low as 0.6 mEq/L, the best results have been obtained with higher levels, such as 0.9 mEq/L.

Drug Interactions Renal clearance of lithium is reduced about 25% by diuretics (eg, thiazides), and doses may need to be reduced by a similar amount. A similar reduction in lithium clearance has been noted with several of the newer nonsteroidal anti-inflammatory drugs that block synthesis of prostaglandins. This interaction has not been reported for either aspirin or acetaminophen. All neuroleptics tested to date, with the possible exception of clozapine and the newer atypical antipsychotics, may produce more severe extrapyramidal syndromes when combined with lithium.

Adverse Effects & Complications Many adverse effects associated with lithium treatment occur at varying times after treatment is started. Some are harmless, but it is important to be alert to adverse effects that may signify impending serious toxic reactions. A. Neurologic and Psychiatric Adverse Effects Tremor is one of the most common adverse effects of lithium treatment, and it occurs with therapeutic doses. Propranolol and atenolol, which have been reported to be effective in essential tremor, also alleviate lithium-induced tremor. Other reported neurologic abnormalities include choreoathetosis, motor hyperactivity, ataxia, dysarthria, and aphasia. Psychiatric disturbances at toxic concentrations are generally marked by mental confusion and withdrawal. Appearance of any new neurologic or psychiatric symptoms or signs is a clear indication for temporarily stopping treatment with lithium and for close monitoring of serum levels.


B. Decreased Thyroid Function Lithium probably decreases thyroid function in most patients exposed to the drug, but the effect is reversible or nonprogressive. Few patients develop frank thyroid enlargement, and fewer still show symptoms of hypothyroidism. Although initial thyroid testing followed by regular monitoring of thyroid function has been proposed, such procedures are not cost-effective. Obtaining a serum TSH concentration every 6–12 months, however, is prudent. C. Nephrogenic Diabetes Insipidus and Other Renal Adverse Effects Polydipsia and polyuria are common but reversible concomitants of lithium treatment, occurring at therapeutic serum concentrations. The principal physiologic lesion involved is loss of responsiveness to antidiuretic hormone (nephrogenic diabetes insipidus). Lithium-induced diabetes insipidus is resistant to vasopressin but responds to amiloride. Extensive literature has accumulated concerning other forms of renal dysfunction during long-term lithium therapy, including chronic interstitial nephritis and minimal-change glomerulopathy with nephrotic syndrome. Some instances of decreased glomerular filtration rate have been encountered but no instances of marked azotemia or renal failure. Patients receiving lithium should avoid dehydration and the associated increased concentration of lithium in urine. Periodic tests of renal concentrating ability should be performed to detect changes. D. Edema Edema is a common adverse effect of lithium treatment and may be related to some effect of lithium on sodium retention. Although weight gain may be expected in patients who become edematous, water retention does not account for the weight gain observed in up to 30% of patients taking lithium. E. Cardiac Adverse Effects The bradycardia-tachycardia (“sick sinus”) syndrome is a definite contraindication to the use of lithium because the ion further depresses the sinus node. T-wave flattening is often observed on the electrocardiogram but is of questionable significance. F. Use During Pregnancy Renal clearance of lithium increases during pregnancy and reverts to lower levels immediately after delivery. A patient whose serum lithium concentration is in a good therapeutic range during pregnancy may develop toxic levels after delivery. Special care in monitoring lithium levels is needed at these times. Lithium is transferred to nursing infants through breast milk, in which it has a concentration about one third to one half that of serum. Lithium toxicity in newborns is manifested by lethargy, cyanosis, poor suck and Moro reflexes, and perhaps hepatomegaly. The issue of lithium-induced dysmorphogenesis is not settled. An earlier report suggested an increase in cardiac anomalies— especially Ebstein’s anomaly—in lithium babies, and it is listed as such in Table 59–1 in this book. However, more recent data suggest that lithium carries a relatively low risk of teratogenic effects. Further research is needed in this important area. G. Miscellaneous Adverse Effects Transient acneiform eruptions have been noted early in lithium treatment. Some of them subside with temporary discontinuance of treatment and do not recur with its resumption. Folliculitis is less dramatic and probably occurs more frequently. Leukocytosis is always present during lithium treatment, probably reflecting a direct effect on leukopoiesis rather than mobilization from the marginal pool. This adverse effect has now become a therapeutic effect in patients with low leukocyte counts.

Overdoses Therapeutic overdoses of lithium are more common than those due to deliberate or accidental ingestion of the drug. Therapeutic overdoses are usually due to accumulation of lithium resulting from some change in the patient’s status, such as diminished serum sodium, use of diuretics, or fluctuating renal function. Since the tissues will have already equilibrated with the blood, the plasma concentrations of lithium may not be excessively high in proportion to the degree of toxicity; any value over 2 mEq/L must be considered as indicating likely toxicity. Because lithium is a small ion, it is dialyzed readily. Both peritoneal dialysis and hemodialysis are effective, although the latter is preferred.

VALPROIC ACID Valproic acid (valproate), discussed in detail in Chapter 24 as an antiepileptic, has been demonstrated to have antimanic effects and is now being widely used for this indication in the USA. (Gabapentin is not effective, leaving the mechanism of antimanic action of


valproate unclear.) Overall, valproic acid shows efficacy equivalent to that of lithium during the early weeks of treatment. It is significant that valproic acid has been effective in some patients who have failed to respond to lithium. For example, mixed states and rapid cycling forms of bipolar disorder may be more responsive to valproate than to lithium. Moreover, its side-effect profile is such that one can rapidly increase the dosage over a few days to produce blood levels in the apparent therapeutic range, with nausea being the only limiting factor in some patients. The starting dosage is 750 mg/d, increasing rapidly to the 1500–2000 mg range with a recommended maximum dosage of 60 mg/kg/d. Combinations of valproic acid with other psychotropic medications likely to be used in the management of either phase of bipolar illness are generally well tolerated. Valproic acid is an appropriate first-line treatment for mania, although it is not clear that it will be as effective as lithium as a maintenance treatment in all subsets of patients. Many clinicians advocate combining valproic acid and lithium in patients who do not fully respond to either agent alone.

CARBAMAZEPINE Carbamazepine has been considered to be a reasonable alternative to lithium when the latter is less than optimally efficacious. However, the pharmacokinetic interactions of carbamazepine and its tendency to induce the metabolism of CYP3A4 substrates make it a more difficult drug to use with other standard treatments for bipolar disorder. The mode of action of carbamazepine is unclear, and oxcarbazepine is not effective. Carbamazepine may be used to treat acute mania and also for prophylactic therapy. Adverse effects (discussed in Chapter 24) are generally no greater and sometimes less than those associated with lithium. Carbamazepine may be used alone or, in refractory patients, in combination with lithium or, rarely, valproate. The use of carbamazepine as a mood stabilizer is similar to its use as an anticonvulsant (see Chapter 24). Dosage usually begins with 200 mg twice daily, with increases as needed. Maintenance dosage is similar to that used for treating epilepsy, ie, 800–1200 mg/d. Plasma concentrations between 3 and 14 mg/L are considered desirable, although no therapeutic range has been established. Blood dyscrasias have figured prominently in the adverse effects of carbamazepine when it is used as an anticonvulsant, but they have not been a major problem with its use as a mood stabilizer. Overdoses of carbamazepine are a major emergency and should generally be managed like overdoses of tricyclic antidepressants (see Chapter 58).

OTHER DRUGS Lamotrigine is approved as a maintenance treatment for bipolar disorder. Although not effective in treating acute mania, it appears effective in reducing the frequency of recurrent depressive cycles and may have some utility in the treatment of bipolar depression. A number of novel agents are under investigation for bipolar depression, including riluzole, a neuroprotective agent that is approved for use in amyotrophic lateral sclerosis; ketamine, a noncompetitive NMDA antagonist previously discussed as a drug believed to model schizophrenia but thought to act by producing relative enhancement of AMPA receptor activity; and AMPA receptor potentiators.

SUMMARY Antipsychotic Drugs & Lithium



PREPARATIONS AVAILABLE



REFERENCES Antipsychotic Drugs Bhattacharjee J, El-Sayeh HG: Aripiprazole versus typical antipsychotic drugs for schizophrenia. Cochrane Database Syst Rev 2008;16;(3):CD006617. Caccia S et al: A new generation of antipsychotics: Pharmacology and clinical utility of cariprazine in schizophrenia. T her Clin Risk Manag 2013;9:319. Chue P: Glycine reuptake inhibition as a new therapeutic approach in schizophrenia: Focus on the glycine transporter 1 (GlyT 1). Curr Pharm Des 2013;19:1311. Citrome L: Cariprazine in bipolar disorder: Clinical efficacy, tolerability, and place in therapy. Adv T her 2013 Feb;30:102. Citrome L: Cariprazine in schizophrenia: Clinical efficacy, tolerability, and place in therapy. Adv T her 2013 Feb;30:114. Citrome L: Cariprazine: Chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability. Expert Opin Drug Metab T oxicol 2013 Feb;9:193. Citrome L: A review of the pharmacology, efficacy and tolerability of recently approved and upcoming oral antipsychotics: An evidence-based medicine approach. CNS Drugs 2013;27:879. Coyle JT : Glutamate and schizophrenia: Beyond the dopamine hypothesis. Cell Mol Neurobiol 2006;26:365. Escamilla MA, Zavala JM: Genetics of bipolar disorder. Dialogues Clin Neurosci 2008;10:141. Fountoulakis KN, Vieta E: T reatment of bipolar disorder: A systematic review of available data and clinical perspectives. Int J Neuropsychopharmacol 2008;11:999. Freudenreich O, Goff DC: Antipsychotic combination therapy in schizophrenia: A review of efficacy and risks of current combinations. Acta Psychiatr Scand 2002;106:323. Glassman AH: Schizophrenia, antipsychotic drugs, and cardiovascular disease. J Clin Psychiatry 2005;66(Suppl 6):5. Grunder G, Nippius H, Carlsson A: T he ‘atypicality’ of antipsychotics: A concept re-examined and re-defined. Nat Rev Drug Discov 2009;8:197. Haddad PM, Anderson IM: Antipsychotic-related QT c prolongation, torsade de pointes and sudden death. Drugs 2002;62:1649. Harrison PJ, Weinberger DR: Schizophrenia genes, gene expression, and neuropathology: On the matter of their convergence. Mol Psychiatry 2005;10:40. Hashimoto K et al: Glutamate modulators as potential therapeutic drugs in schizophrenia and affective disorders. Eur Arch Psychiatry Clin Neurosci 2013;263:367. Herman EJ et al: Metabotropic glutamate receptors for new treatments in schizophrenia. Handb Exp Pharmacol 2012;213:297. Hovelsø N et al: T herapeutic potential of metabotropic glutamate receptor modulators. Curr Neuropharmacol 2012;10:12. Javitt DC: Glycine transport inhibitors in the treatment of schizophrenia. Handb Exp Pharmacol 2012;213:367. Karam CS et al: Signaling pathways in schizophrenia: Emerging targets and therapeutic strategies. T rend Pharmacol Sci 2010;31:381. Lieberman JA et al: Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 2005;353:1209. Lieberman JA et al: Antipsychotic drugs: Comparison in animal models of efficacy, neurotransmitter regulation, and neuroprotection. Pharmacol Rev 2008;60:358. McKeage K, Plosker GL: Amisulpride: A review of its use in the management of schizophrenia. CNS Drugs 2004;18:933. Meltzer HY: T reatment of schizophrenia and spectrum disorders: Pharmacotherapy, psychosocial treatments, and neurotransmitter interactions. Biol Psychiatry 1999;46:1321. Meltzer HY, Massey BW: T he role of serotonin receptors in the action of atypical antipsychotic drugs. Curr Opin Pharmacol 2011;11:59. Meltzer HY et al: A randomized, double-blind comparison of clozapine and high-dose olanzapine in treatment-resistant patients with schizophrenia. J Clin Psychiatry 2008;69:274. Newcomer JW, Haupt DW: T he metabolic effects of antipsychotic medications. Can J Psychiatry 2006;51:480. Schwarz C et al: Valproate for schizophrenia. Cochrane Database Syst Rev 2008;(3):CD004028. Urichuk L et al: Metabolism of atypical antipsychotics: Involvement of cytochrome p450 enzymes and relevance for drug-drug interactions. Curr Drug Metab 2008;9:410. Walsh T et al: Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia. Science 2008;320:539. Zhang A, Neumeyer JL, Baldessarini RJ: Recent progress in development of dopamine receptor subtype-selective agents: Potential therapeutics for neurological and psychiatric disorders. Chem Rev 2007;107:274.

Mood Stabilizers Baraban JM, Worley PF, Snyder SH: Second messenger systems and psychoactive drug action: Focus on the phosphoinositide system and lithium. Am J Psychiatry 1989;146:1251. Bowden CL, Singh V: Valproate in bipolar disorder: 2000 onwards. Acta Psychiatr Scand Suppl 2005;(426):13. Catapano LA, Manji HK: Kinases as drug targets in the treatment of bipolar disorder. Drug Discov T oday 2008;13:295. Fountoulakis KN, Vieta E: T reatment of bipolar disorder: A systematic review of available data and clinical perspectives. Int J Neuropsychopharmacol 2008;11:999. Jope RS: Anti-bipolar therapy: Mechanism of action of lithium. Mol Psychiatry 1999;4:117. Mathew SJ, Manji HK, Charney DS: Novel drugs and therapeutic targets for severe mood disorders. Neuropsychopharmacology 2008;33:2080. Quiroz JA et al: Emerging experimental therapeutics for bipolar disorder: Clues from the molecular pathophysiology. Mol Psychiatry 2004;9:756. Vieta E, Sanchez-Moreno J: Acute and long-term treatment of mania. Dialogues Clin Neurosci 2008;10:165. Yatham LN et al: T hird generation anticonvulsants in bipolar disorder: a review of efficacy and summary of clinical recommendations. J Clin Psychiatry 2002;63:275.

CASE STUDY ANSWER* Schizophrenia is characterized by a disintegration of thought processes and emotional responsiveness. Symptoms commonly include auditory hallucinations, paranoid or bizarre delusions, disorganized thinking and speech, and social and occupational dysfunction. For many patients, typical (eg, haloperidol) and atypical agents (eg, risperidone) are of equal efficacy for treating positive symptoms. Atypical agents are often more effective for treating negative symptoms and cognitive dysfunction and have lower risk of tardive


dyskinesia and hyperprolactinemia. Other indications for the use of selected antipsychotics include bipolar disorder, psychotic depression, Tourette’s syndrome, disturbed behavior in patients with Alzheimer’s disease and in the case of older drugs (eg, chlorpromazine), treatment of emesis and pruritus. * Case Study Answer contributed by A.J. T revor.


_______________ * T he author thanks Herbert Meltzer, MD, PhD, for his contributions to prior editions of this chapter.


CHAPTER

30 Antidepressant Agents Charles DeBattista, MD

CASE STUDY A 47-year-old woman presents to her primary care physician with a chief complaint of fatigue. She indicates that she was promoted to senior manager in her company approximately 11 months earlier. Although her promotion was welcome and came with a sizable raise in pay, it resulted in her having to move away from an office and group of colleagues she very much enjoyed. In addition, her level of responsibility increased dramatically. The patient reports that for the last 7 weeks, she has been waking up at 3 AM every night and been unable to go back to sleep. She dreads the day and the stresses of the workplace. As a consequence, she is not eating as well as she might and has dropped 7% of her body weight in the last 3 months. She also reports being so stressed that she breaks down crying in the office occasionally and has been calling in sick frequently. When she comes home, she finds she is less motivated to attend to chores around the house and has no motivation, interest, or energy to pursue recreational activities that she once enjoyed such as hiking. She describes herself as “chronically miserable and worried all the time.” Her medical history is notable for chronic neck pain from a motor vehicle accident for which she is being treated with tramadol and meperidine. In addition, she is on hydrochlorothiazide and propranolol for hypertension. The patient has a history of one depressive episode after a divorce that was treated successfully with fluoxetine. Medical workup including complete blood cell count, thyroid function tests, and a chemistry panel reveals no abnormalities. She is started on fluoxetine for a presumed major depressive episode and referred for cognitive behavioral psychotherapy. What CYP450 and pharmacodynamic interactions might be associated with fluoxetine use in this patient? Which class of antidepressants would be contraindicated in this patient?

The diagnosis of depression still rests primarily on the clinical interview. Major depressive disorder (MDD) is characterized by depressed mood most of the time for at least 2 weeks or loss of interest or pleasure in most activities, or both. In addition, depression is characterized by disturbances in sleep and appetite as well as deficits in cognition and energy. Thoughts of guilt, worthlessness, and suicide are common. Coronary artery disease, diabetes, and stroke appear to be more common in depressed patients, and depression may considerably worsen the prognosis for patients with a variety of comorbid medical conditions. According to the Centers for Disease Control, antidepressants are consistently among the three most commonly prescribed classes of medications in the USA. The wisdom of such widespread use of antidepressants is debated. However, it is clear that American physicians have been increasingly inclined to use antidepressants to treat a host of conditions and that patients have been increasingly receptive to their use. The primary indication for antidepressant agents is the treatment of MDD. Major depression, with a lifetime prevalence of around 17% in the USA and a point prevalence of 5%, is associated with substantial morbidity and mortality. MDD represents one of the most common causes of disability in the developed world. In addition, major depression is commonly associated with a variety of medical conditions—from chronic pain to coronary artery disease. When depression coexists with other medical conditions, the patient’s disease burden increases, and the quality of life—and often the prognosis for effective treatment—decreases significantly. Some of the growth in antidepressant use may be related to the broad application of these agents for conditions other than major depression. For example, antidepressants have received FDA approvals for the treatment of panic disorder, generalized anxiety disorder (GAD), post-traumatic stress disorder (PTSD), and obsessive-compulsive disorder (OCD). In addition, antidepressants are commonly used to treat pain disorders such as neuropathic pain and the pain associated with fibromyalgia. Some antidepressants are used for treating premenstrual dysphoric disorder (PMDD), mitigating the vasomotor symptoms of menopause, and treating stress urinary incontinence. Thus, antidepressants have a broad spectrum of use in medical practice. However, their primary use remains the treatment for MDD.


Pathophysiology of Major Depression There has been a marked shift in the last decade in our understanding of the pathophysiology of major depression. In addition to the older idea that a deficit in function or amount of monoamines (the monoamine hypothesis) is central to the biology of depression, there is evidence that neurotrophic and endocrine factors play a major role (the neurotrophic hypothesis). Histologic studies, structural and functional brain imaging research, genetic findings, and steroid research all suggest a complex pathophysiology for MDD with important implications for drug treatment.

Neurotrophic Hypothesis There is substantial evidence that nerve growth factors such as brain-derived neurotrophic factor (BDNF) are critical in the regulation of neural plasticity, resilience, and neurogenesis. The evidence suggests that depression is associated with the loss of neurotrophic support and that effective antidepressant therapies increase neurogenesis and synaptic connectivity in cortical areas such as the hippocampus. BDNF is thought to exert its influence on neuronal survival and growth effects by activating the tyrosine kinase receptor B in both neurons and glia (Figure 30–1).

FIGURE 30–1 The neurotrophic hypothesis of major depression. Changes in trophic factors (especially brain-derived neurotrophic


factor, BDNF) and hormones appear to play a major role in the development of major depression (A). Successful treatment results in changes in these factors (B). CREB, cAMP response element-binding (protein). BDNF, brain-derived neurotrophic factor. (Reproduced, with permission, from Nestler EJ: Neurobiology of depression. Neuron 2002;34[1]:13–25. Copyright Elsevier.) Several lines of evidence support the neurotrophic hypothesis. Animal and human studies indicate that stress and pain are associated with a drop in BDNF levels and that this loss of neurotrophic support contributes to atrophic structural changes in the hippocampus and perhaps other areas such as the medial frontal cortex and anterior cingulate. The hippocampus is known to be important both in contextual memory and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Likewise, the anterior cingulate plays a role in the integration of emotional stimuli and attention functions, whereas the medial orbital frontal cortex is also thought to play a role in memory, learning, and emotion. Over 30 structural imaging studies suggest that major depression is associated with a 5–10% loss of volume in the hippocampus, although some studies have not replicated this finding. Depression and chronic stress states have also been associated with a substantial loss of volume in the anterior cingulate and medial orbital frontal cortex. Loss of volume in structures such as the hippocampus also appears to increase as a function of the duration of illness and the amount of time that the depression remains untreated. Another source of evidence supporting the neurotrophic hypothesis of depression comes from studies of the direct effects of BDNF on emotional regulation. Direct infusion of BDNF into the midbrain, hippocampus, and lateral ventricles of rodents has an antidepressantlike effect in animal models. Moreover, all known classes of antidepressants are associated with an increase in BDNF levels in animal models with chronic (but not acute) administration. This increase in BDNF levels is consistently associated with increased neurogenesis in the hippocampus in these animal models. Other interventions thought to be effective in the treatment of major depression, including electroconvulsive therapy, also appear to robustly stimulate BDNF levels and hippocampus neurogenesis in animal models. Human studies seem to support the animal data on the role of neurotrophic factors in stress states. Depression appears to be associated with a drop in BDNF levels in the cerebrospinal fluid and serum as well as with a decrease in tyrosine kinase receptor B activity. Conversely, administration of antidepressants increases BDNF levels in clinical trials and may be associated with an increase in hippocampus volume in some patients. Much evidence supports the neurotrophic hypothesis of depression, but not all evidence is consistent with this concept. Animal studies in BDNF knockout mice have not always suggested an increase in depressive or anxious behaviors that would be expected with a deficiency of BDNF. In addition, some animal studies have found an increase in BDNF levels after some types of social stress and an increase rather than a decrease in depressive behaviors with lateral ventricle injections of BDNF. A proposed explanation for the discrepant findings on the role of neurotrophic factors in depression is that there are polymorphisms for BDNF that may yield very different effects. Mutations in the BDNF gene have been found to be associated with altered anxiety and depressive behavior in both animal and human studies. Thus, the neurotrophic hypothesis continues to be intensely investigated and has yielded new insights and potential targets in the treatment of MDD.

Monoamines & Other Neurotransmitters The monoamine hypothesis of depression (Figure 30–2) suggests that depression is related to a deficiency in the amount or function of cortical and limbic serotonin (5-HT), norepinephrine (NE), and dopamine (DA).



FIGURE 30–2 The amine hypothesis of major depression. Depression appears to be associated with changes in serotonin or norepinephrine signaling in the brain (or both) with significant downstream effects. Most antidepressants cause changes in amine signaling. AC, adenylyl cyclase; 5-HT, serotonin; CREB, cAMP response element-binding (protein); DAG, diacyl glycerol; IP 3 , inositol trisphosphate; MAO, monoamine oxidase; NET, norepinephrine transporter; PKC, protein kinase C; PLC, phospholipase C; SERT, serotonin transporter. (Adapted from Belmaker R, Agam G: Major depressive disorder. N Engl J Med 2008;358:59.) Evidence to support the monoamine hypothesis comes from several sources. It has been known for many years that reserpine treatment, which is known to deplete monoamines, is associated with depression in a subset of patients. Similarly, depressed patients who respond to serotonergic antidepressants such as fluoxetine often rapidly suffer relapse when given diets free of tryptophan, a precursor of serotonin synthesis. Patients who respond to noradrenergic antidepressants such as desipramine are less likely to relapse on a tryptophan-free diet. Moreover, depleting catecholamines in depressed patients who have previously responded to noradrenergic agents likewise tends to be associated with relapse. Administration of an inhibitor of norepinephrine synthesis is also associated with a rapid return of depressive symptoms in patients who respond to noradrenergic but not necessarily in patients who had responded to serotonergic antidepressants. Another line of evidence supporting the monoamine hypothesis comes from genetic studies. A functional polymorphism exists for the promoter region of the serotonin transporter gene, which regulates how much of the transporter protein is available. Subjects who are homozygous for the s (short) allele may be more vulnerable to developing major depression and suicidal behavior in response to stress. In addition, homozygotes for the s allele may also be less likely to respond to and tolerate serotonergic antidepressants. Conversely, subjects with the l (long) allele tend to be more resistant to stress and may be more likely to respond to serotonergic antidepressants. Studies of depressed patients have sometimes shown an alteration in monoamine function. For example, some studies have found evidence of alteration in serotonin receptor numbers (5-HT1A and 5-HT2C) or norepinephrine (ι2 ) receptors in depressed and suicidal patients, but these findings have not been consistent. A reduction in the primary serotonin metabolite 5-hydroxyindoleacetic acid in the cerebrospinal fluid is associated with violent and impulsive behavior, including violent suicide attempts. However, this finding is not specific to major depression and is associated more generally with violent and impulsive behavior. Finally, perhaps the most convincing line of evidence supporting the monoamine hypothesis is the fact that (at the time of this writing) all available antidepressants appear to have significant effects on the monoamine system. All classes of antidepressants appear to enhance the synaptic availability of 5-HT, norepinephrine, or dopamine. Attempts to develop antidepressants that work on other neurotransmitter systems have not been effective to date. The monoamine hypothesis, like the neurotrophic hypothesis, is at best incomplete. Many studies have not found an alteration in function or levels of monoamines in depressed patients. In addition, some candidate antidepressant agents under study do not act directly on the monoamine system. In addition to the monoamines, the excitatory neurotransmitter glutamate appears to be important in the pathophysiology of depression. A number of studies of depressed patients have found elevated glutamate content in the cerebrospinal fluid of depressed patients and decreased glutamine/glutamate ratios in their plasma. In addition, postmortem studies have revealed significant increases in the frontal and dorsolateral prefrontal cortex of depressed patients. Likewise, structural neuroimaging studies have consistently found volumetric changes in the brain areas of depressed patients in which glutamate neurons and their connections are most abundant, including the amygdala and hippocampus. Antidepressants are known to impact glutamate neurotransmission in a variety of ways. For example, chronic antidepressant use is associated with reducing glutamatergic transmission, including the presynaptic release of glutamate in the hippocampus and cortical areas. Similarly, the chronic administration of antidepressants significantly reduces depolarization-evoked release of glutamate in animal models. Stress is known to enhance the release of glutamate in rodents, and antidepressants inhibit stress-induced presynaptic release of glutamate in these models. Given the effect of antidepressants on the glutamate system, there has been a growing interest in the development of pharmaceutical agents that might modulate the glutamate system. Ketamine is a potent, high-affinity, noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that has long been used in anesthesia and is a common drug of abuse in some parts of the world. A number of preclinical and clinical studies have demonstrated rapid antidepressant effects of ketamine. Multiple studies have suggested that a single dose of intravenous ketamine at subanesthetic doses produces rapid relief of depression, even in treatment-resistant patients, that may persist for 1 week or longer. Unfortunately, ketamine is associated with cognitive, dissociative, and psychotomimetic properties that make it impractical as a long-term treatment for depression. Still, a number of other NMDA receptor antagonists, partial antagonists, and metabotropic glutamate receptor modulators (see Chapter 29) are under investigation as potential antidepressants.

Neuroendocrine Factors in the Pathophysiology of Depression Depression is known to be associated with a number of hormonal abnormalities. Among the most replicated of these findings are abnormalities in the HPA axis in patients with MDD. Moreover, MDD is associated with elevated cortisol levels ( Figure 30–1), nonsuppression of adrenocorticotropic hormone (ACTH) release in the dexamethasone suppression test, and chronically elevated levels of corticotropin-releasing hormone. The significance of these HPA abnormalities is unclear, but they are thought to indicate a


dysregulation of the stress hormone axis. More severe types of depression, such as psychotic depression, tend to be associated with HPA abnormalities more commonly than milder forms of major depression. It is well known that both exogenous glucocorticoids and endogenous elevation of cortisol are associated with mood symptoms and cognitive deficits similar to those seen in MDD. Thyroid dysregulation has also been reported in depressed patients. Up to 25% of depressed patients are reported to have abnormal thyroid function. These abnormalities include a blunting of response of thyrotropin to thyrotropin-releasing hormone, and elevations in circulating thyroxine during depressed states. Clinical hypothyroidism often presents with depressive symptoms, which resolve with thyroid hormone supplementation. Thyroid hormones are also commonly used in conjunction with standard antidepressants to augment therapeutic effects of the latter. Finally, sex steroids are also implicated in the pathophysiology of depression. Estrogen deficiency states, which occur in the postpartum and postmenopausal periods, are thought to play a role in the etiology of depression in some women. Likewise, severe testosterone deficiency in men is sometimes associated with depressive symptoms. Hormone replacement therapy in hypogonadal men and women may be associated with an improvement in mood and depressive symptoms.

Integration of Hypotheses Regarding the Pathophysiology of Depression The several pathophysiologic hypotheses just described are not mutually exclusive. It is evident that the monoamine, neuroendocrine, and neurotrophic systems are interrelated in important ways. For example, HPA and steroid abnormalities may contribute to suppression of transcription of the BDNF gene. Glucocorticoid receptors are found in high density in the hippocampus. Binding of these hippocampal glucocorticoid receptors by cortisol during chronic stress states such as major depression may decrease BDNF synthesis and may result in volume loss in stress-sensitive regions such as the hippocampus. The chronic activation of monoamine receptors by antidepressants appears to have the opposite effect of stress and results in an increase in BDNF transcription. In addition, activation of monoamine receptors appears to down-regulate the HPA axis and may normalize HPA function. One of the weaknesses of the monoamine hypothesis is the fact that amine levels increase immediately with antidepressant use, but maximum beneficial effects of most antidepressants are not seen for many weeks. The time required to synthesize neurotrophic factors has been proposed as an explanation for this delay of antidepressant effects. Appreciable protein synthesis of products such as BDNF typically takes 2 weeks or longer and coincides with the clinical course of antidepressant treatment.

BASIC PHARMACOLOGY OF ANTIDEPRESSANTS Chemistry & Subgroups The currently available antidepressants make up a remarkable variety of chemical types. These differences and the differences in their molecular targets provide the basis for distinguishing several subgroups. A. Selective Serotonin Reuptake Inhibitors The selective serotonin reuptake inhibitors (SSRIs) represent a chemically diverse class of agents that have as their primary action the inhibition of the serotonin transporter (SERT; Figure 30–3). Fluoxetine was introduced in the United States in 1988 and quickly became one of the most commonly prescribed medications in medical practice. The development of fluoxetine emerged out of the search for chemicals that had high affinity for monoamine receptors but lacked the affinity for histamine, acetylcholine, and ι adrenoceptors that is seen with the tricyclic antidepressants (TCAs). There are currently six available SSRIs, and they are the most common antidepressants in clinical use. In addition to their use in major depression, SSRIs have indications in GAD, PTSD, OCD, panic disorder, PMDD, and bulimia. Fluoxetine, sertraline, and citalopram exist as isomers and are formulated in the racemic forms, whereas paroxetine and fluvoxamine are not optically active. Escitalopram is the (S) enantiomer of citalopram. As with all antidepressants, SSRIs are highly lipophilic. The popularity of SSRIs stems largely from their ease of use, safety in overdose, relative tolerability, cost (all are available as generic products), and broad spectrum of uses.


FIGURE 30–3 Structures of several selective serotonin reuptake inhibitors (SSRIs). B. Serotonin-Norepinephrine Reuptake Inhibitors Two classes of antidepressants act as combined serotonin and norepinephrine reuptake inhibitors: selective serotonin-norepinephrine reuptake inhibitors (SNRIs) and TCAs. 1. Selective serotonin-norepinephrine reuptake inhibitors—The SNRIs include venlafaxine, its metabolite desvenlafaxine, duloxetine, and levomilnacipran. Levomilnacipran is the active enantiomer of a racemic SNRI, milnacipran. Milnacipran has been approved for the treatment of fibromyalgia in the USA and has been used in the treatment of depression in Europe for many years. In addition to their use in major depression, SNRIs have applications in the treatment of pain disorders including neuropathies and fibromyalgia. SNRIs are also used in the treatment of generalized anxiety, stress urinary incontinence, and vasomotor symptoms of menopause.


SNRIs are chemically unrelated to each other. Venlafaxine was discovered in the process of evaluating chemicals that inhibit binding of imipramine. Venlafaxine’s in vivo effects are similar to those of imipramine but with a more favorable adverse-effect profile. All SNRIs bind the serotonin (SERT) and norepinephrine (NET) transporters, as do the TCAs. However, unlike the TCAs, the SNRIs do not have much affinity for other receptors. Venlafaxine and desvenlafaxine are bicyclic compounds, whereas duloxetine is a three-ring structure unrelated to the TCAs. Milnacipran contains a cyclopropane ring and is provided as a racemic mixture.

2. Tricyclic antidepressants—The TCAs were the dominant class of antidepressants until the introduction of SSRIs in the 1980s and 1990s. Nine TCAs are available in the USA, and they all have an iminodibenzyl (tricyclic) core (Figure 30–4). The chemical differences between the TCAs are relatively subtle. For example, the prototype TCA imipramine and its metabolite, desipramine, differ by only a methyl group in the propylamine side chain. However, this minor difference results in a substantial change in their pharmacologic profiles. Imipramine is highly anticholinergic and is a relatively strong serotonin as well as norepinephrine reuptake inhibitor. In contrast, desipramine is much less anticholinergic and is a more potent and somewhat more selective norepinephrine reuptake inhibitor than is imipramine.


FIGURE 30–4 Structures of some tricyclic antidepressants (TCAs). At the present time, the TCAs are used primarily in depression that is unresponsive to more commonly used antidepressants such as the SSRIs or SNRIs. Their loss of popularity stems in large part from relatively poorer tolerability compared with newer agents, difficulty of use, and lethality in overdose. Other uses for TCAs include the treatment of pain conditions, enuresis, and insomnia. C. 5-HT2 Receptor Modulators Two antidepressants are thought to act primarily as antagonists at the 5-HT 2 receptor: trazodone and nefazodone. Trazodone’s structure includes a triazolo moiety that is thought to impart antidepressant effects. Its primary metabolite, m-chlorphenylpiperazine (mcpp), is a potent 5-HT2 antagonist. Trazodone was among the most commonly prescribed antidepressants until it was supplanted by the SSRIs in the late 1980s. The most common use of trazodone in current practice is as an unlabeled hypnotic, since it is highly sedating and not associated with tolerance or dependence.


Nefazodone is chemically related to trazodone. Its primary metabolites, hydroxynefazodone and m-cpp are both inhibitors of the 5HT2 receptor. Nefazodone received an FDA black box warning in 2001 implicating it in hepatotoxicity, including lethal cases of hepatic failure. Though still available generically, nefazodone is no longer commonly prescribed. The primary indications for both nefazodone and trazodone are major depression, although both have also been used in the treatment of anxiety disorders.

Vortioxetine is a newer agent that acts as an antagonist of the 5-HT 3 , 5-HT7 , and 5-HT1D receptors, a partial agonist of the 5-HT1B receptor, and an agonist of the 5HT 1A receptor. It also inhibits the serotonin transporter but its actions are not primarily related to SERT inhibition and it is therefore not classified as an SSRI. Vortioxetine has demonstrated efficacy on major depression in a number of controlled clinical studies. In addition, there is some preliminary evidence that the drug also may improve some aspects of cognition in depressed patients. D. Tetracyclic and Unicyclic Antidepressants A number of antidepressants do not fit neatly into the other classes. Among these are bupropion, mirtazapine, amoxapine, vilazodone, and maprotiline (Figure 30–5). Bupropion has a unicyclic aminoketone structure. Its unique structure results in a different side-effect profile than most antidepressants (described below). Bupropion somewhat resembles amphetamine in chemical structure and, like the stimulant, has central nervous system (CNS) activating properties.


FIGURE 30–5 Structures of the tetracyclics, amoxapine, maprotiline, and mirtazapine and the unicyclic, bupropion. Mirtazapine was introduced in 1994 and, like bupropion, is one of the few antidepressants not commonly associated with sexual effects. It has a tetracyclic chemical structure and belongs to the piperazino-azepine group of compounds. Mirtazapine, amoxapine, and maprotiline have tetracyclic structures. Amoxapine is the N-methylated metabolite of loxapine, an older antipsychotic drug. Amoxapine and maprotiline share structural similarities and side effects comparable to the TCAs. As a result, these tetracyclics are not commonly prescribed in current practice. Their primary use is in MDD that is unresponsive to other agents. Vilazodone has a multi-ring structure that allows it to bind potently to the serotonin transporter but minimally to the dopamine and norepinephrine transporter. E. Monoamine Oxidase Inhibitors Arguably the first modern class of antidepressants, monoamine oxidase inhibitors (MAOIs) were introduced in the 1950s but are now rarely used in clinical practice because of toxicity and potentially lethal food and drug interactions. Their primary use now is in the treatment of depression unresponsive to other antidepressants. However, MAOIs have also been used historically to treat anxiety states, including social anxiety and panic disorder. In addition, selegiline is used in the treatment of Parkinson’s disease (see Chapter 28). Current MAOIs include the hydrazine derivatives phenelzine and isocarboxazid and the non-hydrazines tranylcypromine, selegiline, and moclobemide (the latter is not available in the USA). The hydrazines and tranylcypromine bind irreversibly and nonselectively with MAO-A and -B, whereas other MAOIs may have more selective or reversible properties. Some of the MAOIs such as tranylcypromine resemble amphetamine in chemical structure, whereas other MAOIs such as selegiline have amphetamine-like metabolites. As a result, these MAOIs tend to have substantial CNS-stimulating effects.


Pharmacokinetics The antidepressants share several pharmacokinetic features (Table 30–1). Most have fairly rapid oral absorption, achieve peak plasma levels within 2–3 hours, are tightly bound to plasma proteins, undergo hepatic metabolism, and are renally cleared. However, even within classes, the pharmacokinetics of individual antidepressants varies considerably. TABLE 30–1 Pharmacokinetic profiles of selected antidepressants.



A. Selective Serotonin Reuptake Inhibitors The prototype SSRI, fluoxetine, differs from other SSRIs in some important respects (Table 30–1). Fluoxetine is metabolized to an active product, norfluoxetine, which may have plasma concentrations greater than those of fluoxetine. The elimination half-life of norfluoxetine is about three times longer than fluoxetine and contributes to the longest half-life of all the SSRIs. As a result, fluoxetine has to be discontinued 4 weeks or longer before an MAOI can be administered to mitigate the risk of serotonin syndrome. Fluoxetine and paroxetine are potent inhibitors of the CYP2D6 isoenzyme, and this contributes to potential drug interactions (see Drug Interactions). In contrast, fluvoxamine is an inhibitor of CYP3A4, whereas citalopram, escitalopram, and sertraline have more modest CYP interactions. B. Serotonin-Norepinephrine Reuptake Inhibitors 1. Selective serotonin-norepinephrine reuptake inhibitors—Venlafaxine is extensively metabolized in the liver via the CYP2D6 isoenzyme to O-desmethylvenlafaxine (desvenlafaxine). Both have similar half-lives of about 8-11 hours. Despite the relatively short half-lives, both drugs are available in formulations that allow once-daily dosing. Venlafaxine and desvenlafaxine have the lowest protein binding of all antidepressants (27–30%). Unlike most antidepressants, desvenlafaxine is conjugated and does not undergo extensive oxidative metabolism. At least 45% of desvenlafaxine is excreted unchanged in the urine compared with 4–8% of venlafaxine. Duloxetine is well absorbed and has a half-life of 12-15 hours but is dosed once daily. It is tightly bound to protein (97%) and undergoes extensive oxidative metabolism via CYP2D6 and CYP1A2. Hepatic impairment significantly alters duloxetine levels unlike desvenlafaxine. Both milnacipran and levomilnacipran are well absorbed after oral dosing.Both have shorter half-lives and lower protein binding than venlafaxine (Table 30–1). Milnacipran and levomilnacipran are largely excreted unchanged in the urine. Levomilnacipran also undergoes desethylation via 3A3/4. 2. Tricyclic antidepressants—The TCAs tend to be well absorbed and have long half-lives (Table 30–1). As a result, most are dosed once daily at night because of their sedating effects. TCAs undergo extensive metabolism via demethylation, aromatic hydroxylation, and glucuronide conjugation. Only about 5% of TCAs are excreted unchanged in the urine. The TCAs are substrates of the CYP2D6 system, and the serum levels of these agents tend to be substantially influenced by concurrent administration of drugs such as fluoxetine. In addition, genetic polymorphism for CYP2D6 may result in low or extensive metabolism of the TCAs. The secondary amine TCAs, including desipramine and nortriptyline, lack active metabolites and have fairly linear kinetics. These TCAs have a wide therapeutic window, and serum levels are reliable in predicting response and toxicity. C. 5-HT Receptor Modulators Trazodone and nefazodone are rapidly absorbed and undergo hepatic metabolism. Both drugs are bound to protein and have limited bioavailability because of extensive metabolism. Because of their short half-lives split dosing is generally required when these drugs are used as antidepressants. However, trazodone is often prescribed as a single dose at night as a hypnotic in lower doses than are used in the treatment of depression. Both trazodone and nefazodone have active metabolites that also exhibit 5-HT2 antagonism. Nefazodone is a potent inhibitor of the CYP3A4 system and may interact with drugs metabolized by this enzyme (see Drug Interactions). Vortioxetine is not a potent inhibitor of CYP isoenzymes. However, it is extensively metabolized through oxidation by CYP2D6 and other isoenzymes and then undergoes subsequent glucuronic acid conjugation. It is tightly bound to protein and has linear and dose-proportional pharmacokinetics. D. Tetracyclic and Unicyclic Agents Bupropion is rapidly absorbed and has a mean protein binding of 85%. It undergoes extensive hepatic metabolism and has a substantial first-pass effect. It has three active metabolites including hydroxybupropion; the latter is being developed as an antidepressant. Bupropion has a biphasic elimination with the first phase lasting about 1 hour and the second phase lasting 14 hours. Amoxapine is also rapidly absorbed with protein binding of about 85%. The half-life is variable, and the drug is often given in divided doses. Amoxapine undergoes extensive hepatic metabolism. One of the active metabolites, 7-hydroxyamoxapine, is a potent D 2 blocker and is associated with antipsychotic effects. Maprotiline is similarly well absorbed orally and 88% bound to protein. It undergoes extensive hepatic metabolism. Mirtazapine is demethylated followed by hydroxylation and glucuronide conjugation. Several CYP isozymes are involved in the metabolism of mirtazapine, including 2D6, 3A4, and 1A2. The half-life of mirtazapine is 20–40 hours, and it is usually dosed once in the evening because of its sedating effects. Vilazodone is well absorbed (Table 30–1) and absorption is increased when it is given with a fatty meal. It is extensively metabolized by CYP3A4 with minor contributions by CYP2C19 and CYP2D6. Only 1% of vilazodone is excreted unchanged in the urine. E. Monoamine Oxidase Inhibitors


The different MAOIs are metabolized via different pathways but tend to have extensive first-pass effects that may substantially decrease bioavailability. Tranylcypromine is ring hydroxylated and N-acetylated, whereas acetylation appears to be a minor pathway for phenelzine. Selegiline is N-demethylated and then hydroxylated. The MAOIs are well absorbed from the gastrointestinal tract. Because of the prominent first-pass effects and their tendency to inhibit MAO in the gut (resulting in tyramine pressor effects), alternative routes of administration are being developed. For example, selegiline is available in both transdermal and sublingual forms that bypass both gut and liver. These routes decrease the risk of food interactions and provide substantially increased bioavailability.

Pharmacodynamics As previously noted, all currently available antidepressants enhance monoamine neurotransmission by one of several mechanisms. The most common mechanism is inhibition of the activity of SERT, NET, or both monoamine transporters ( Table 30–2). Antidepressants that inhibit SERT, NET, or both include the SSRIs and SNRIs (by definition), and the TCAs. Another mechanism for increasing the availability of monoamines is inhibition of their enzymatic degradation (by the MAOIs). Additional strategies for enhancing monoamine tone include binding presynaptic autoreceptors (mirtazapine) or specific postsynaptic receptors (5-HT2 antagonists and mirtazapine). Ultimately, the increased availability of monoamines for binding in the synaptic cleft results in a cascade of events that enhance the transcription of some proteins and the inhibition of others. It is the net production of these proteins, including BDNF, glucocorticoid receptors, β adrenoceptors, and other proteins, that appears to determine the benefits as well as the toxicity of a given agent. TABLE 30–2 Antidepressant effects on several receptors and transporters.



A. Selective Serotonin Reuptake Inhibitors The serotonin transporter (SERT) is a glycoprotein with 12 transmembrane regions embedded in the axon terminal and cell body membranes of serotonergic neurons. When extracellular serotonin binds to receptors on the transporter, conformational changes occur in the transporter and serotonin, Na+, and Cl- are moved into the cell. Binding of intracellular K+ then results in the release of serotonin inside the cell and return of the transporter to its original conformation. SSRIs allosterically inhibit the transporter by binding the SERT receptor at a site other than the serotonin binding site. At therapeutic doses, about 80% of the activity of the transporter is inhibited. Functional polymorphisms exist for SERT that determine the activity of the transporter (Table 30–2). SSRIs have modest effects on other neurotransmitters. Unlike TCAs and SNRIs, there is little evidence that SSRIs have prominent effects on β adrenoceptors or the norepinephrine transporter, NET. Binding to the serotonin transporter is associated with tonic inhibition of the dopamine system, although there is substantial interindividual variability in this effect. The SSRIs do not bind aggressively to histamine, muscarinic, or other receptors. B. Drugs That Block Both Serotonin and Norepinephrine Transporters A large number of antidepressants have mixed inhibitory effects on both serotonin and norepinephrine transporters. The newer agents in this class (venlafaxine and duloxetine) are termed SNRIs; those in the older group are termed TCAs on the basis of their structures. 1. Serotonin-norepinephrine reuptake inhibitors—SNRIs bind both the serotonin and the norepinephrine transporters. The NET is structurally very similar to the 5-HT transporter. Like the serotonin transporter, it is a 12-transmembrane domain complex that allosterically binds norepinephrine. The NET also has a moderate affinity for dopamine. Venlafaxine is a weak inhibitor of NET, whereas desvenlafaxine, duloxetine, milnacipran, and levomilnacipran are more balanced inhibitors of both SERT and NET. Nonetheless, the affinity of most SNRIs tends to be much greater for SERT than for NET. The SNRIs differ from the TCAs in that they lack the potent antihistamine, α-adrenergic blocking, and anticholinergic effects of the TCAs. As a result, the SNRIs tend to be favored over the TCAs in the treatment of MDD and pain syndromes because of their better tolerability. 2. Tricyclic antidepressants—The TCAs resemble the SNRIs in function, and their antidepressant activity is thought to relate primarily to their inhibition of 5-HT and norepinephrine reuptake. Within the TCAs, there is considerable variability in affinity for SERT versus NET. For example, clomipramine has relatively very little affinity for NET but potently binds SERT. This selectivity for the serotonin transporter contributes to clomipramine’s known benefits in the treatment of OCD. On the other hand, the secondary amine TCAs, desipramine and nortriptyline, are relatively more selective for NET. Although the tertiary amine TCA imipramine has more serotonin effect initially, its metabolite, desipramine, then balances this effect with more NET inhibition. Common adverse effects of the TCAs, including dry mouth and constipation, are attributable to the potent antimuscarinic effects of many of these drugs. The TCAs also tend to be potent antagonists of the histamine H1 receptor. TCAs such as doxepin are sometimes prescribed as hypnotics and used in treatments for pruritus because of their antihistamine properties. The blockade of α adrenoceptors can result in substantial orthostatic hypotension, particularly in older patients. C. 5-HT Receptor Modulators The principle action of both nefazodone and trazodone appears to be blockade of the 5-HT2A receptor. Inhibition of this receptor in both animal and human studies is associated with substantial antianxiety, antipsychotic, and antidepressant effects. Conversely, agonists of the 5-HT2A receptor, eg, lysergic acid (LSD) and mescaline, are often hallucinogenic and anxiogenic. The 5-HT 2A receptor is a G proteincoupled receptor and is distributed throughout the neocortex. Nefazodone is a weak inhibitor of both SERT and NET but is a potent antagonist of the postsynaptic 5-HT 2A receptor, as are its metabolites. Trazodone is also a weak but selective inhibitor of SERT with little effect on NET. Its primary metabolite, m-cpp, is a potent 5-HT2 antagonist, and much of trazodone’s benefits as an antidepressant might be attributed to this effect. Trazodone also has weak-tomoderate presynaptic α-adrenergic–blocking properties and is a modest antagonist of the H1 receptor. As described above, vortioxetine has multimodal effects on a variety of 5-HT receptors and is an allosteric inhibitor of SERT. It has no known direct activity on norepinephrine or dopamine receptors. D. Tetracyclic and Unicyclic Antidepressants The actions of bupropion remain poorly understood. Bupropion and its major metabolite hydroxybupropion are modest-to-moderate inhibitors of norepinephrine and dopamine reuptake in animal studies. However, these effects seem less than are typically associated with antidepressant benefit. A more significant effect of bupropion is presynaptic release of catecholamines. In animal studies, bupropion appears to substantially increase the presynaptic availability of norepinephrine, and dopamine to a lesser extent. Bupropion has virtually no direct effects on the serotonin system. Mirtazapine has a complex pharmacology. It is an antagonist of the presynaptic α 2 autoreceptor and enhances the release of both


norepinephrine and 5-HT. In addition, mirtazapine is an antagonist of 5-HT 2 and 5-HT3 receptors. Finally, mirtazapine is a potent H 1 antagonist, which is associated with the drug’s sedative effects. The actions of amoxapine and maprotiline resemble those of TCAs such as desipramine. Both are potent NET inhibitors and less potent SERT inhibitors. In addition, both possess anticholinergic properties. Unlike the TCAs or other antidepressants, amoxapine is a moderate inhibitor of the postsynaptic D2 receptor. As such, amoxapine possesses some antipsychotic properties. Vilazodone is a potent serotonin reuptake inhibitor and a partial agonist of the 5-HT 1A receptor. Partial agonists of the 5-HT 1A receptor such as buspirone are thought to have mild to moderate antidepressant and anxiolytic properties. E. Monoamine Oxidase Inhibitors MAOIs act by mitigating the actions of monoamine oxidase in the neuron and increasing monoamine content. There are two forms of monoamine oxidase. MAO-A is present in both dopamine and norepinephrine neurons and is found primarily in the brain, gut, placenta, and liver; its primary substrates are norepinephrine, epinephrine, and serotonin. MAO-B is found primarily in serotonergic and histaminergic neurons and is distributed in the brain, liver, and platelets. MAO-B acts primarily on dopamine, tyramine, phenylethylamine, and benzylamine. Both MAO-A and -B metabolize tryptamine. MAOIs are classified by their specificity for MAO-A or -B and whether their effects are reversible or irreversible. Phenelzine and tranylcypromine are examples of irreversible, nonselective MAOIs. Moclobemide is a reversible and selective inhibitor of MAO-A but is not available in the USA. Moclobemide can be displaced from MAO-A by tyramine, and this mitigates the risk of food interactions. In contrast, selegiline is an irreversible MAO-B–specific agent at low doses. Selegiline is useful in the treatment of Parkinson’s disease at these low doses, but at higher doses it becomes a nonselective MAOI similar to other agents.

CLINICAL PHARMACOLOGY OF ANTIDEPRESSANTS Clinical Indications A. Depression The FDA indication for the use of the antidepressants in the treatment of major depression is fairly broad. Most antidepressants are approved for both acute and long-term treatment of major depression. Acute episodes of MDD tend to last about 6–14 months untreated, but at least 20% of episodes last 2 years or longer. The goal of acute treatment of MDD is remission of all symptoms. Since antidepressants may not achieve their maximum benefit for 1–2 months or longer, it is not unusual for a trial of therapy to last 8–12 weeks at therapeutic doses. The antidepressants are successful in achieving remission in about 30–40% of patients within a single trial of 8–12 weeks. If an inadequate response is obtained, therapy is often switched to another agent or augmented by addition of another drug. For example, bupropion, an atypical antipsychotic, or mirtazapine might be added to an SSRI or SNRI to augment antidepressant benefit if monotherapy is unsuccessful. Seventy to eighty percent of patients are able to achieve remission with sequenced augmentation or switching strategies. Once an adequate response is achieved, continuation therapy is recommended for a minimum of 6–12 months to reduce the substantial risk of relapse. Approximately 85% of patients who have a single episode of MDD will have at least one recurrence in a lifetime. Many patients have multiple recurrences, and these recurrences may progress to more serious, chronic, and treatment-resistant episodes. Thus, it is not unusual for patients to require maintenance treatment to prevent recurrences. Although maintenance treatment studies of more than 5 years are uncommon, long-term studies with TCAs, SNRIs, and SSRIs suggest a significant protective benefit when given chronically. Thus, it is commonly recommended that patients be considered for long-term maintenance treatment if they have had two or more serious MDD episodes in the previous 5 years or three or more serious episodes in a lifetime. It is not clear whether antidepressants are useful for all subtypes of depression. For example, patients with bipolar depression may not benefit much from antidepressants even when added to mood stabilizers. In fact, the antidepressants are sometimes associated with switches into mania or more rapid cycling. There has also been some debate about the overall efficacy of antidepressants in unipolar depression, with some meta-analyses showing large effects and others showing more modest effects. Although this debate is not likely to be settled immediately, there is little debate that antidepressants have important benefits for most patients. Psychotherapeutic interventions such as cognitive behavioral therapy appear to be as effective as antidepressant treatment for mild to moderate forms of depression. However, cognitive behavioral therapy tends to take longer to be effective and is generally more expensive than antidepressant treatment. Psychotherapy is often combined with antidepressant treatment, and the combination appears more effective than either strategy alone. B. Anxiety Disorders After major depression, anxiety disorders represent the most common application of antidepressants. A number of SSRIs and SNRIs have been approved for all the major anxiety disorders, including PTSD, OCD, social anxiety disorder, GAD, and panic disorder. Panic disorder is characterized by recurrent episodes of brief overwhelming anxiety, which often occur without precipitant. Patients may begin to fear having an attack, or they avoid situations in which they might have an attack. In contrast, GAD is characterized by a chronic,


free-floating anxiety and undue worry that tends to be chronic in nature. Although older antidepressants and drugs of the sedativehypnotic class are still occasionally used for the treatment of anxiety disorders, SSRIs and SNRIs have largely replaced them. The benzodiazepines (see Chapter 22) provide much more rapid relief of both generalized anxiety and panic than do any of the antidepressants. However, the antidepressants appear to be at least as effective as, and perhaps more effective than, benzodiazepines in the long-term treatment of these anxiety disorders. Furthermore, antidepressants do not carry the risks of dependence and tolerance that may occur with the benzodiazepines. OCD is known to respond to serotonergic antidepressants. It is characterized by repetitive anxiety-provoking thoughts (obsessions) or repetitive behaviors aimed at reducing anxiety (compulsions). Clomipramine and several of the SSRIs are approved for the treatment of OCD, and they are moderately effective. Behavior therapy is usually combined with the antidepressant for additional benefits. Social anxiety disorder is an uncommonly diagnosed but a fairly common condition in which patients experience severe anxiety in social interactions. This anxiety may limit their ability to function adequately in their jobs or interpersonal relationships. Several SSRIs and venlafaxine are approved for the treatment of social anxiety. The efficacy of the SSRIs in the treatment of social anxiety is greater in some studies than their efficacy in the treatment of MDD. PTSD is manifested when a traumatic or life-threatening event results in intrusive anxiety-provoking thoughts or imagery, hypervigilance, nightmares, and avoidance of situations that remind the patient of the trauma. SSRIs are considered first-line treatment for PTSD and can benefit a number of symptoms including anxious thoughts and hypervigilance. Other treatments, including psychotherapeutic interventions, are usually required in addition to antidepressants. C. Pain Disorders It has been known for over 40 years that antidepressants possess analgesic properties independent of their mood effects. TCAs have been used in the treatment of neuropathic and other pain conditions since the 1960s. Medications that possess both norepinephrine and 5HT reuptake blocking properties are often useful in treating pain disorders. Ascending corticospinal monoamine pathways appear to be important in the endogenous analgesic system. In addition, chronic pain conditions are commonly associated with major depression. TCAs continue to be commonly used for some of these conditions, and SNRIs are increasingly used. In 2010, duloxetine was approved for the treatment of chronic joint and muscle pain. As mentioned earlier, milnacipran is approved for the treatment of fibromyalgia in the USA and for MDD in other countries. Other SNRIs, eg, desvenlafaxine, are being investigated for a variety of pain conditions from postherpetic neuralgia to chronic back pain. D. Premenstrual Dysphoric Disorder Approximately 5% of women in the child-bearing years will have prominent mood and physical symptoms during the late luteal phase of almost every cycle; these may include anxiety, depressed mood, irritability, insomnia, fatigue, and a variety of other physical symptoms. These symptoms are more severe than those typically seen in premenstrual syndrome (PMS) and can be quite disruptive to vocational and interpersonal activities. The SSRIs are known to be beneficial to many women with PMDD, and fluoxetine and sertraline are approved for this indication. Treating for 2 weeks out of the month in the luteal phase may be as effective as continuous treatment. The rapid effects of SSRIs in PMDD may be associated with rapid increases in pregnenolone levels. E. Smoking Cessation Bupropion was approved in 1997 as a treatment for smoking cessation. Approximately twice as many people treated with bupropion as with placebo have a reduced urge to smoke. In addition, patients taking bupropion appear to experience fewer mood symptoms and possibly less weight gain while withdrawing from nicotine dependence. Bupropion appears to be about as effective as nicotine patches in smoking cessation. The mechanism by which bupropion is helpful in this application is unknown, but the drug may mimic nicotine’s effects on dopamine and norepinephrine and may antagonize nicotinic receptors. Nicotine is also known to have antidepressant effects in some people, and bupropion may substitute for this effect. Other antidepressants may also have a role in the treatment of smoking cessation. Nortriptyline has been shown to be helpful in smoking cessation, but the effects have not been as consistent as those seen with bupropion. F. Eating Disorders Bulimia nervosa and anorexia nervosa are potentially devastating disorders. Bulimia is characterized by episodic intake of large amounts of food (binges) followed by ritualistic purging through emesis, the use of laxatives, or other methods. Medical complications of the purging, such as hypokalemia, are common and dangerous. Anorexia is a disorder in which reduced food intake results in a loss of weight of 15% or more of ideal body weight, and the person has a morbid fear of gaining weight and a highly distorted body image. Anorexia is often chronic and may be fatal in 10% or more of cases. Antidepressants appear to be helpful in the treatment of bulimia but not anorexia. Fluoxetine was approved for the treatment of bulimia in 1996, and other antidepressants have shown benefit in reducing the binge-purge cycle. The primary treatment for anorexia at this time is refeeding, family therapy, and cognitive behavioral therapy. Bupropion may have some benefits in treating obesity. Nondepressed, obese patients treated with bupropion were able to lose


somewhat more weight and maintain the loss relative to a similar population treated with placebo. However, the weight loss was not robust, and there appear to be more effective options for weight loss. G. Other Uses for Antidepressants Antidepressants are used for many other on- and off-label applications. Enuresis in children is an older labeled use for some TCAs, but they are less commonly used now because of their side effects. The SNRI duloxetine is approved in Europe for the treatment of urinary stress incontinence. Many of the serotonergic antidepressants appear to be helpful for treating vasomotor symptoms in perimenopause. Desvenlafaxine is under consideration for FDA approval for the treatment of these vasomotor symptoms, and studies have suggested that SSRIs, venlafaxine, and nefazodone may also provide benefit. Although serotonergic antidepressants are commonly associated with inducing sexual adverse effects, some of these effects might prove useful for some sexual disorders. For example, SSRIs are known to delay orgasm in some patients. For this reason, SSRIs are sometimes used to treat premature ejaculation. In addition, bupropion has been used to treat sexual adverse effects associated with SSRI use, although its efficacy for this use has not been consistently demonstrated in controlled trials.

Choosing an Antidepressant The choice of an antidepressant depends first on the indication. Not all conditions are equally responsive to all antidepressants. However, in the treatment of MDD, it is difficult to demonstrate that one antidepressant is consistently more effective than another. Thus, the choice of an antidepressant for the treatment of depression rests primarily on practical considerations such as cost, availability, adverse effects, potential drug interactions, the patient’s history of response or lack thereof, and patient preference. Other factors such as the patient’s age, gender, and medical status may also guide antidepressant selection. For example, older patients are particularly sensitive to the anticholinergic effects of the TCAs. On the other hand, the CYP3A4-inhibiting effects of the SSRI fluvoxamine may make this a problematic choice in some older patients because fluvoxamine may interact with many other medications that an older patient may require. There is some suggestion that female patients may respond to and tolerate serotonergic better than noradrenergic or TCA antidepressants, but the data supporting this gender difference have not been consistent. Patients with narrow-angle glaucoma may have an exacerbation with noradrenergic antidepressants, whereas bupropion and other antidepressants are known to lower the seizure threshold in epilepsy patients. At present, SSRIs are the most commonly prescribed first-line agents in the treatment of both MDD and anxiety disorders. Their popularity comes from their ease of use, tolerability, and safety in overdose. The starting dose of the SSRIs is usually the same as the therapeutic dose for most patients, and so titration may not be required. In addition, most SSRIs are now generically available and inexpensive. Other agents, including the SNRIs, bupropion, and mirtazapine, are also reasonable first-line agents for the treatment of MDD. Bupropion, mirtazapine, and nefazodone are the antidepressants with the least association with sexual side effects and are often prescribed for this reason. However, bupropion is not thought to be effective in the treatment of the anxiety disorders and may be poorly tolerated in anxious patients. The primary indication for bupropion is in the treatment of major depression, including seasonal (winter) depression. Off-label uses of bupropion include the treatment of attention deficit hyperkinetic disorder (ADHD), and bupropion is commonly combined with other antidepressants to augment therapeutic response. The primary indication for mirtazapine is in the treatment of major depression. However, its strong antihistamine properties have contributed to its occasional use as a hypnotic and as an adjunctive treatment to more activating antidepressants. The TCAs and MAOIs are now relegated to second- or third-line treatments for MDD. Both the TCAs and the MAOIs are potentially lethal in overdose, require titration to achieve a therapeutic dose, have serious drug interactions, and have many troublesome adverse effects. As a consequence, their use in the treatment of MDD or anxiety is now reserved for patients who have been unresponsive to other agents. Clearly, there are patients whose depression responds only to MAOIs or TCAs. Thus, TCAs and MAOIs are probably underused in treatment-resistant depressed patients. The use of antidepressants outside the treatment of MDD tends to require specific agents. For example, the TCAs and SNRIs appear to be useful in the treatment of pain conditions, but other antidepressant classes appear to be far less effective. SSRIs and the highly serotonergic TCA, clomipramine, are effective in the treatment of OCD, but noradrenergic antidepressants have not proved to be as helpful for this condition. Bupropion and nortriptyline have usefulness in the treatment of smoking cessation, but SSRIs have not been proven useful. Thus, outside the treatment of depression, the choice of antidepressant is primarily dependent on the known benefit of a particular antidepressant or class for a particular indication.

Dosing The optimal dose of an antidepressant depends on the indication and on the patient. For SSRIs, SNRIs, and a number of newer agents, the starting dose for the treatment of depression is usually a therapeutic dose (Table 30–3). Patients who show little or no benefit after at least 4 weeks of treatment may benefit from a higher dose even though it has been difficult to show a clear advantage for higher doses with SSRIs, SNRIs, and other newer antidepressants. The dose is generally titrated to the maximum dosage recommended or to the highest dosage tolerated if the patient is not responsive to lower doses. Some patients may benefit from doses lower than the usual


minimum recommended therapeutic dose. TCAs and MAOIs typically require titration to a therapeutic dosage over several weeks. Dosing of the TCAs may be guided by monitoring TCA serum levels. TABLE 30–3 Antidepressant dose ranges.



Some anxiety disorders may require higher doses of antidepressants than are used in the treatment of major depression. For example, patients treated for OCD often require maximum or somewhat higher than maximum recommended MDD doses to achieve optimal benefits. Likewise, the minimum dose of paroxetine for the effective treatment of panic disorder is higher than the minimum dose required for the effective treatment of depression. In the treatment of pain disorders, modest doses of TCAs are often sufficient. For example, 25–50 mg/d of imipramine might be beneficial in the treatment of pain associated with a neuropathy, but this would be a subtherapeutic dose in the treatment of MDD. In contrast, SNRIs are usually prescribed in pain disorders at the same doses used in the treatment of depression.

Adverse Effects Although some potential adverse effects are common to all antidepressants, most of their adverse effects are specific to a subclass of agents and to their pharmacodynamic effects. An FDA warning applied to all antidepressants is the risk of increased suicidality in patients younger than 25. The warning suggests that use of antidepressants is associated with suicidal ideation and gestures, but not completed suicides, in up to 4% of patients under 25 who were prescribed antidepressant in clinical trials. This rate is about twice the rate seen with placebo treatment. For those over 25, there is either no increased risk or a reduced risk of suicidal thoughts and gestures on antidepressants, particularly after age 65. Although a small minority of patients may experience a treatment-emergent increase in suicidal ideation with antidepressants, the absence of treatment of a major depressive episode in all age groups is a particularly important risk factor in completed suicides. A. Selective Serotonin Reuptake Inhibitors The adverse effects of the most commonly prescribed antidepressants—the SSRIs—can be predicted from their potent inhibition of SERT. SSRIs enhance serotonergic tone, not just in the brain but throughout the body. Increased serotonergic activity in the gut is commonly associated with nausea, gastrointestinal upset, diarrhea, and other gastrointestinal symptoms. Gastrointestinal adverse effects usually emerge early in the course of treatment and tend to improve after the first week. Increasing serotonergic tone at the level of the spinal cord and above is associated with diminished sexual function and interest. As a result, at least 30–40% of patients treated with SSRIs report loss of libido, delayed orgasm, or diminished arousal. The sexual effects often persist as long as the patient remains on the antidepressant but may diminish with time. Other adverse effects related to the serotonergic effects of SSRIs and vortioxetine include an increase in headaches and insomnia or hypersomnia. Some patients gain weight while taking SSRIs, particularly paroxetine. Sudden discontinuation of short half-life SSRIs such as paroxetine and sertraline is associated with a discontinuation syndrome in some patients characterized by dizziness, paresthesias, and other symptoms beginning 1 or 2 days after stopping the drug and persisting for 1 week or longer. Most antidepressants are category C agents by the FDA teratogen classification system. There is an association of paroxetine with cardiac septal defects in first trimester exposures. Thus, paroxetine is a category D agent. Other possible associations of SSRIs with post-birth complications, including pulmonary hypertension, have not been clearly established. B. Serotonin-Norepinephrine Reuptake Inhibitors and Tricyclic Antidepressants SNRIs have many of the serotonergic adverse effects associated with SSRIs. In addition, SNRIs may also have noradrenergic effects, including increased blood pressure and heart rate, and CNS activation, such as insomnia, anxiety, and agitation. The hemodynamic effects of SNRIs tend not to be problematic in most patients. A dose-related increase in blood pressure has been seen more commonly with the immediate-release form of venlafaxine than with other SNRIs. Likewise, there are more reports of cardiac toxicity with venlafaxine overdose than with either the other SNRIs or SSRIs. Duloxetine is rarely associated with hepatic toxicity in patients with a history of liver damage. All the SNRIs have been associated with a discontinuation syndrome resembling that seen with SSRI discontinuation. The primary adverse effects of TCAs have been described in the previous text. Anticholinergic effects are perhaps the most common. These effects result in dry mouth, constipation, urinary retention, blurred vision, and confusion. They are more common with tertiary amine TCAs such as amitriptyline and imipramine than with the secondary amine TCAs desipramine and nortriptyline. The potent α-blocking property of TCAs often results in orthostatic hypotension. H1 antagonism by the TCAs is associated with weight gain and sedation. The TCAs are class 1A antiarrhythmic agents (see Chapter 14) and are arrhythmogenic at higher doses. Sexual effects are common, particularly with highly serotonergic TCAs such as clomipramine. The TCAs have a prominent discontinuation syndrome characterized by cholinergic rebound and flulike symptoms. C. 5-HT Receptor Modulators The most common adverse effects associated with the 5-HT2 antagonists are sedation and gastrointestinal disturbances. Sedative effects, particularly with trazodone, can be quite pronounced. Thus, it is not surprising that the treatment of insomnia is currently the primary application of trazodone. The gastrointestinal effects appear to be dose-related and are less pronounced than those seen with SNRIs or SSRIs. Sexual effects are uncommon with nefazodone or trazodone treatment as a result of the relatively selective


serotonergic effects of these drugs on the 5-HT2 receptor rather than on SERT. However, trazodone has rarely been associated with inducing priapism. Both nefazodone and trazodone are Îą-blocking agents and may result in a dose-related orthostatic hypotension in some patients. Nefazodone has been associated with hepatotoxicity, including rare fatalities and cases of fulminant hepatic failure requiring transplantation. The rate of serious hepatoxicity with nefazodone has been estimated at 1 in 250,000 to 1 in 300,000 patient-years of nefazodone treatment. As with the SSRIs, the most common adverse effects of vortioxetine are serotonergic and include dose-dependent gastrointestinal effects, particularly nausea, as well as sexual dysfunction. Higher doses of vortioxetine tend to increase the rate of GI and sexual side effects. The teratogenic risks of vortioxetine are not known but like most other antidepressants, it is considered a category C agent. D. Tetracyclics and Unicyclics Amoxapine is sometimes associated with a parkinsonian syndrome due to its D2 -blocking action. Mirtazapine has significant sedative effect. Maprotiline has a moderately high affinity for NET and may cause TCA-like adverse effects and, rarely, seizures. Bupropion is occasionally associated with agitation, insomnia, and anorexia. Vilazodone may have somewhat higher rates of gastrointestinal upset, including diarrhea and nausea, than the SSRIs. E. Monoamine Oxidase Inhibitors The most common adverse effects of the MAOIs leading to discontinuation of these drugs are orthostatic hypotension and weight gain. In addition, the irreversible nonselective MAOIs are associated with the highest rates of sexual effects of all the antidepressants. Anorgasmia is fairly common with therapeutic doses of some MAOIs. The amphetamine-like properties of some MAOIs contributes to activation, insomnia, and restlessness in some patients. Phenelzine tends to be more sedating than either selegiline or tranylcypromine. Confusion is also sometimes associated with higher doses of MAOIs. Because they block metabolism of tyramine and similar ingested amines, MAOIs may cause dangerous interactions with certain foods and with serotonergic drugs (see Interactions). Finally, MAOIs have been associated with a sudden discontinuation syndrome manifested in a delirium-like presentation with psychosis, excitement, and confusion.

Overdose Suicide attempts are a common and unfortunate consequence of major depression. The lifetime risk of completing suicide in patients previously hospitalized with MDD may be as high as 15%. Overdose is the most common method used in suicide attempts, and antidepressants, especially the TCAs, are frequently involved. Overdose can induce lethal arrhythmias, including ventricular tachycardia and fibrillation. In addition, blood pressure changes and anticholinergic effects including altered mental status and seizures are sometimes seen in TCA overdoses. A 1500 mg dose of imipramine or amitriptyline (less than 7 days’ supply at antidepressant doses) is enough to be lethal in many patients. Toddlers taking 100 mg will likely show evidence of toxicity. Treatment typically involves cardiac monitoring, airway support, and gastric lavage. Sodium bicarbonate is often administered to displace the TCA from cardiac sodium channels. An overdose with an MAOI can produce a variety of effects including autonomic instability, hyperadrenergic symptoms, psychotic symptoms, confusion, delirium, fever, and seizures. Management of MAOI overdoses usually involves cardiac monitoring, vital signs support, and lavage. Compared with TCAs and MAOIs, the other antidepressants are generally much safer in overdose. Fatalities with SSRI overdose alone are extremely uncommon. Similarly, SNRIs tend to be much safer in overdose than the TCAs. However, venlafaxine has been associated with some cardiac toxicity in overdose and appears to be less safe than SSRIs. Bupropion is associated with seizures in overdose, and mirtazapine may be associated with sedation, disorientation, and tachycardia. With the newer agents, fatal overdoses often involve the combination of the antidepressant with other drugs, including alcohol. Management of overdose with the newer antidepressants usually involves emptying of gastric contents and vital sign support as the initial intervention.

Drug Interactions Antidepressants are commonly prescribed with other psychotropic and nonpsychotropic agents. There is potential for drug interactions with all antidepressants, but the most serious of these involve the MAOIs and to a lesser extent the TCAs. A. Selective Serotonin Reuptake Inhibitors The most common interactions with SSRIs are pharmacokinetic interactions. For example, paroxetine and fluoxetine are potent CYP2D6 inhibitors (Table 30–4). Thus, administration with 2D6 substrates such as TCAs can lead to dramatic and sometimes unpredictable elevations in the tricyclic drug concentration. The result may be toxicity from the TCA. Similarly, fluvoxamine, a CYP3A4 inhibitor, may elevate the levels of concurrently administered substrates for this enzyme such as diltiazem and induce bradycardia or hypotension. Other SSRIs, such as citalopram and escitalopram, are relatively free of pharmacokinetic interactions. The most serious interaction with the SSRIs are pharmacodynamic interactions with MAOIs that produce a serotonin syndrome (see below).


TABLE 30–4 Some antidepressant–CYP450 drug interactions.

B. Selective Serotonin-Norepinephrine Reuptake Inhibitors and Tricyclic Antidepressants The SNRIs have relatively fewer CYP450 interactions than the SSRIs. Venlafaxine is a substrate but not an inhibitor of CYP2D6 or other isoenzymes, whereas desvenlafaxine is a minor substrate for CYP3A4. Duloxetine is a moderate inhibitor of CYP2D6 and so may elevate TCA and levels of other CYP2D6 substrates. Since milnacipran is neither a substrate nor potent inducer of CYP450 isoenzymes, is not tightly protein bound, and is largely excreted unchanged in the urine, it is unlikely to have clinically significant pharmacokinetic drug interactions. On the other hand, levomilnacipran is reported to be a substrate of CYP3A4 and the dosage of the drug should be lowered when combined with potent inhibitors of CYP3A4 such as ketoconazole. Like all serotonergic antidepressants, SNRIs are contraindicated in combination with MAOIs. Elevated TCA levels may occur when these drugs are combined with CYP2D6 inhibitors or from constitutional factors. About 7% of the Caucasian population in the USA has a CYP2D6 polymorphism that is associated with slow metabolism of TCAs and other 2D6 substrates. Combination of a known CYP2D6 inhibitor and a TCA in a patient who is a slow metabolizer may result in markedly increased effects. Such an interaction has been implicated, though rarely, in cases of TCA toxicity. There may also be additive anticholinergic or antihistamine effects when TCAs are combined with other agents that share these properties such as benztropine or diphenhydramine. Similarly, antihypertensive drugs may exacerbate the orthostatic hypotension induced by TCAs. C. 5-HT Receptor Modulators Nefazodone is an inhibitor of the CYP3A4 isoenzyme, so it can raise the level and thus exacerbate adverse effects of many 3A4dependent drugs. For example, triazolam levels are increased by concurrent administration of nefazodone such that a reduction in triazolam dosage by 75% is recommended. Likewise, administration of nefazodone with simvastatin has been associated with 20-fold increase in plasma levels of simvastatin. Trazodone is a substrate but not a potent inhibitor of CYP3A4. As a result, combining trazodone with potent inhibitors of CYP3A4, such as ritonavir or ketoconazole, may lead to substantial increases in trazodone levels. Vortioxetine is a substrate of CYP2D6 and 2B6 and it is recommended that the dose be cut in half when it is co-administered with fluoxetine or bupropion. Inducers of CYP isoenzymes such as rifampin, carbamazepine, and phenytoin will lower serum levels of vortioxetine and may require increasing the dose of vortioxetine.


D. Tetracyclic and Unicyclic Antidepressants Bupropion is metabolized primarily by CYP2B6, and its metabolism may be altered by drugs such as cyclophosphamide, which is a substrate of 2B6. The major metabolite of bupropion, hydroxybupropion, is a moderate inhibitor of CYP2D6 and so can raise desipramine levels. Bupropion should be avoided in patients taking MAOIs. Mirtazapine is a substrate for several CYP450 enzymes including 2D6, 3A4, and 1A2. Consequently, drugs that inhibit these isozymes may raise mirtazapine levels. However, mirtazapine is not an inhibitor of these enzymes. The sedating effects of mirtazapine may be additive with those of CNS depressants such as alcohol and benzodiazepines. Amoxapine and maprotiline share most drug interactions common to the TCA group. Both are CYP2D6 substrates and should be used with caution in combination with inhibitors such as fluoxetine. Amoxapine and maprotiline also both have anticholinergic and antihistaminic properties that may be additive with drugs that share a similar profile. Since vilazodone is primarily a substrate of CYP3A4, strong CYP3A4 inhibitors such as ketoconazole can increase the serum concentration of vilazodone by 50% or more. On the other hand, vilazodone is neither a potent inhibitor nor a strong inducer of any CYP isoenzymes. It may be a mild inducer of CYP2C19. E. Monoamine Oxidase Inhibitors MAOIs are associated with two classes of serious drug interactions. The first of these is the pharmacodynamic interaction of MAOIs with serotonergic agents including SSRIs, SNRIs, and most TCAs along with some analgesic agents such as meperidine. These combinations of an MAOI with a serotonergic agent may result in a life-threatening serotonin syndrome (see Chapter 16). The serotonin syndrome is thought to be caused by overstimulation of 5-HT receptors in the central gray nuclei and the medulla. Symptoms range from mild to lethal and include a triad of cognitive (delirium, coma), autonomic (hypertension, tachycardia, diaphoreses), and somatic (myoclonus, hyperreflexia, tremor) effects. Most serotonergic antidepressants should be discontinued at least 2 weeks before starting an MAOI. Fluoxetine, because of its long half-life, should be discontinued for 4–5 weeks before an MAOI is initiated. Conversely, an MAOI must be discontinued for at least 2 weeks before starting a serotonergic agent. The second serious interaction with MAOIs occurs when an MAOI is combined with tyramine in the diet or with sympathomimetic substrates of MAO. An MAOI prevents the breakdown of tyramine in the gut, and this results in high serum levels that enhance peripheral noradrenergic effects, including raising blood pressure dramatically. Patients on an MAOI who ingest large amounts of dietary tyramine may experience malignant hypertension and subsequently a stroke or myocardial infarction. Thus, patients taking MAOIs require a low-tyramine diet and should avoid foods such as aged cheeses, tap beer, soy products, and dried sausages, which contain high amounts of tyramine (see Chapter 9). Similar sympathomimetics also may cause significant hypertension when combined with MAOIs. Thus, over-the-counter cold preparations that contain pseudoephedrine and phenylpropanolamine are contraindicated in patients taking MAOIs.

SUMMARY Antidepressants



PREPARATIONS AVAILABLE

REFERENCES Aan Het Rot M et al: Ketamine for depression: Where do we go from here? Biol Psychiatry 2012;72:537. Alam MY et al: Safety, tolerability, and efficacy of vortioxetine (Lu AA21004) in major depressive disorder: Results of an open-label, flexible-dose, 52-week extension study.


Int Clin Psychopharmacol 2014;29:39. Alessandro S, Kato M: T he serotonin transporter gene and effectiveness of SSRIs. Expert Rev Neurother 2008;8(1):111. Bab I, Yirmiya R: Depression, selective serotonin reuptake inhibitors, and osteoporosis. Curr Osteoporos Rep 2010;8:185. Barrera AZ, T orres LD, Munoz RF: Prevention of depression: T he state of the science at the beginning of the 21st century. Int Rev Psychiatry 2007;19:655. Bellingham GA, Peng PW: Duloxetine: A review of its pharmacology and use in chronic pain management. Reg Anesth Pain Med 2010;35:294. Belmaker R, Agam G: Major depressive disorder. N Engl J Med 2008;358:55. Bockting CL et al: Continuation and maintenance use of antidepressants in recurrent depression. Psychother Psychosom 2008;77:17. Bonisch H, Bruss M: T he norepinephrine transporter in physiology and disease. Handb Exp Pharmacol 2006;175:485. Castren E, Voikar V, Rantamaki T : Role of neurotrophic factors in depression. Curr Opin Pharmacol 2007;7:18. Chaki S et al: mGlu2/3 and mGlu5 receptors: Potential targets for novel antidepressants. Neuropharmacology 2013;66:40. Chappell AS et al: A double-blind, randomized, placebo-controlled study of the efficacy and safety of duloxetine for the treatment of chronic pain due to osteoarthritis of the knee. Pain Pract 2011;11:33. Chen G et al: Pharmacokinetic drug interactions involving vortioxetine (Lu AA21004), a multimodal antidepressant. Clin Drug Investig 2013;33:727. Cipriani A et al: Fluoxetine versus other types of pharmacotherapy for depression. Cochrane Database Syst Rev 2005;(4):CD004185. Cipriani A et al: Metareview on short-term effectiveness and safety of antidepressants for depression: An evidence-based approach to inform clinical practice. Can J Psychiatry 2007;52:553. Citrome L: Vortioxetine for major depressive disorder: A systematic review of the efficacy and safety profile for this newly approved antidepressant - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract 2014;68:60. de Beaurepaire R: Questions raised by the cytokine hypothesis of depression. Brain Behav Immun 2002;16:610. Dhillon S, Scott LJ, Plosker GL: Escitalopram: A review of its use in the management of anxiety disorders. CNS Drugs 2006;20:763. Dhillon S, Yang LP, Curran MP: Bupropion: A review of its use in the management of major depressive disorder. Drugs 2008;68:653. Duman RS, Monteggia LM: A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006;59:1116. Dvir Y, Smallwood P: Serotonin syndrome: A complex but easily avoidable condition. Gen Hosp Psychiatry 2008;30:284. Fontenelle LF et al: An update on the pharmacological treatment of obsessive-compulsive disorder. Expert Opin Pharmacother 2007;8:563. Geisser ME et al: A pooled analysis of two randomized, double-blind, placebo-controlled trials of milnacipran monotherapy in the treatment of fibromyalgia. Pain Pract 2011;11:120. Gether U et al: Neurotransmitter transporters: Molecular function of important drug targets. T rends Pharmacol Sci 2006;27:375. Gillespie CF, Nemeroff CB: Hypercortisolemia and depression. Psychosom Med 2005;67(Suppl 1):S26. Gillman PK: A review of serotonin toxicity data: Implications for the mechanisms of antidepressant drug action. Biol Psychiatry 2006;59:1046. Gillman PK: T ricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br J Pharmacol 2007;151:737. Giner L et al: Selective serotonin reuptake inhibitors and the risk for suicidality in adolescents: An update. Int J Adolesc Med Health 2005;17:211. Guay DR: Vilazodone hydrochloride, a combined SSRI and 5-HT 1A receptor agonist for major depressive disorder. Consult Pharm 2012;27:857. Gutman DA, Owens MJ: Serotonin and norepinephrine transporter binding profile of SSRIs. Essent Psychopharmacol 2006;7:35. Hirschfeld RM: Antidepressants in long-term therapy: A review of tricyclic antidepressants and selective serotonin reuptake inhibitors. Acta Psychiatr Scand Suppl 2000;403:35. Hirschfeld RM: History and evolution of the monoamine hypothesis of depression. J Clin Psychiatry 2000;61(Suppl 6):4. Holma KM et al: Long-term outcome of major depressive disorder in psychiatric patients is variable. J Clin Psychiatry 2008;69:196. Jann MW, Slade JH: Antidepressant agents for the treatment of chronic pain and depression. Pharmacotherapy 2007;27:1571. Kalia M: Neurobiological basis of depression: An update. Metabolism 2005;54(5 Suppl 1):24. Kozisek ME, Middlemas D, Bylund DB: Brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B in the mechanism of action of antidepressant therapies. Pharmacol T her 2008;117:30. Krystal JH, Sanacora G, Duman RS. Rapid-acting glutamatergic antidepressants: T he path to ketamine and beyond. Biol Psychiatry 2013;73:1133. Laughren T P et al: Vilazodone: Clinical basis for the US Food and Drug Administration’s approval of a new antidepressant. J Clin Psychiatry 2011;72:1166. Lesch KP, Gutknecht L: Pharmacogenetics of the serotonin transporter. Prog Neuropsychopharmacol Biol Psychiatry 2005;29:1062. Mago R et al: Safety and tolerability of levomilnacipran ER in major depressive disorder: Results from an open-label, 48-week extension study. Clin Drug Investig 2013;33:761. Maletic V et al: Neurobiology of depression: An integrated view of key findings. Int J Clin Pract 2007;61:2030. Manji HK, Drevets WC, Charney DS: T he cellular neurobiology of depression. Nat Med 2001;7:541. Mathews DC, Zarate CA Jr. Current status of ketamine and related compounds for depression. J Clin Psychiatry 2013;74:516. McCleane G: Antidepressants as analgesics. CNS Drugs 2008;22:139. McEwen BS: Glucocorticoids, depression, and mood disorders: Structural remodeling in the brain. Metabolism 2005;54(5 Suppl 1):20. Moller HJ: Evidence for beneficial effects of antidepressants on suicidality in depressive patients: A systematic review. Eur Arch Psychiatry Clin Neurosci 2006;256:329. Montgomery SA, et al: Efficacy and safety of levomilnacipran sustained release in moderate to severe major depressive disorder: a randomized, double-blind, placebocontrolled, proof-of-concept study. J Clin Psychiatry. 2013;74:363. Nestler EJ et al: Neurobiology of depression. Neuron 2002;34:13. Pace T W, Hu F, Miller AH: Cytokine-effects on glucocorticoid receptor function: Relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun 2007;21:9. Pilc A, Wieronska JM, Skolnick P: Glutamate-based antidepressants: Preclinical psychopharmacology. Biol Psychiatry 2013;73:1125. Sakinofsky I: T reating suicidality in depressive illness. Part 2: Does treatment cure or cause suicidality? Can J Psychiatry 2007;52(6 Suppl 1):85S. Schatzberg AF, Cole JO, DeBattista C: Manual of Clinical Psychopharmacology, 6th ed. American Psychiatric Publishing, 2007. Shapiro JR et al: Bulimia nervosa treatment: A systematic review of randomized controlled trials. Int J Eat Disord 2007;40:321. Soomro GM et al: Selective serotonin reuptake inhibitors (SSRIs) versus placebo for obsessive compulsive disorder (OCD). Cochrane Database Syst Rev 2008;(1):CD001765. Stein MB, Stein DJ: Social anxiety disorder. Lancet 2008;371(9618):1115.


Stone EA, Lin Y, Quartermain D: A final common pathway for depression? Progress toward a general conceptual framework. Neurosci Biobehav Rev 2008;32:508. T uccori M et al: Use of selective serotonin reuptake inhibitors during pregnancy and risk of major and cardiovascular malformations: An update. Postgrad Med 2010;122:49. Warden D et al: T he ST AR* D Project results: A comprehensive review of findings. Curr Psychiatry Rep 2007;9:449. Wheeler BW et al: T he population impact on incidence of suicide and non-fatal self harm of regulatory action against the use of selective serotonin reuptake inhibitors in under 18s in the United Kingdom: Ecological study. Br Med J 2008;336(7643):542. Wilson KL et al: Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol 2011;28:19. Yu S, Holsboer F, Almeida OF: Neuronal actions of glucocorticoids: Focus on depression. J Steroid Biochem Mol Biol 2008;108:300.

CASE STUDY ANSWER Fluoxetine, the prototype SSRI, has a number of pharmacokinetic and pharmacodynamic interactions. Fluoxetine is a CYP450 2D6 inhibitor and thus can inhibit the metabolism of 2D6 substrates such as propranolol and other β blockers; tricyclic antidepressants; tramadol; opioids such as methadone, codeine, and oxycodone; antipsychotics such as haloperidol and thioridazine; and many other drugs. This inhibition of metabolism can result in significantly higher plasma levels of the concurrent drug, and this may lead to an increase in adverse reactions associated with that drug. As a potent inhibitor of the serotonin transporter, fluoxetine is associated with a number of pharmacodynamic interactions involving serotonergic neurotransmission. The combination of tramadol with fluoxetine has occasionally been associated with a serotonin syndrome, characterized by diaphoreses, autonomic instability, myoclonus, seizures, and coma. The combination of fluoxetine with an MAOI is contraindicated because of the risk of a fatal serotonin syndrome. In addition, meperidine is specifically contraindicated in combination with an MAOI.


CHAPTER

31 Opioid Agonists & Antagonists* Mark A. Schumacher, PhD, MD, Allan I. Basbaum, PhD, & Ramana K. Naidu, MD

CASE STUDY A 60-year-old man with a history of moderate chronic obstructive pulmonary disease presents in the emergency department with a broken hip suffered in an automobile accident. He complains of severe pain. What is the most appropriate immediate treatment for his pain? Are any special precautions needed?

Morphine, the prototypic opioid agonist, has long been known to relieve severe pain with remarkable efficacy. The opium poppy is the source of crude opium from which Sertürner in 1803 isolated morphine, the pure alkaloid, naming it after Morpheus, the Greek god of dreams. It remains the standard against which all drugs that have strong analgesic action are compared. These drugs are collectively known as opioids and include not only the natural and semisynthetic alkaloid derivatives from opium but also synthetic surrogates, other opioid-like drugs whose actions are blocked by the nonselective antagonist naloxone, plus several endogenous peptides that interact with the different subtypes of opioid receptors.

BASIC PHARMACOLOGY OF THE OPIOIDS Source Opium, the source of morphine, is obtained from the poppy, Papaver somniferum and P album. After incision, the poppy seed pod exudes a white substance that turns into a brown gum that is crude opium. Opium contains many alkaloids, the principal one being morphine, which is present in a concentration of about 10%. Codeine is synthesized commercially from morphine.

Classification & Chemistry The term opioid describes all compounds that work at opioid receptors. The term opiate specifically describes the naturally occurring alkaloids: morphine, codeine, thebaine, and papaverine. In contrast, narcotic was originally used to describe sleep-inducing medications, but in the United States, its usage has shifted into a legal term. Opioid drugs include full agonists, partial agonists, and antagonists–measures of intrinsic activity or efficacy. Morphine is a full agonist at the l (mu)-opioid receptor, the major analgesic opioid receptor (Table 31–1). Opioids may also differ in receptor binding affinity. For example, morphine exhibits a greater binding affinity at the μ-opioid receptor than does codeine. Other opioid receptor subtypes include c (delta) and j (kappa) receptors. Simple substitution of an allyl group on the nitrogen of the full agonist morphine plus addition of a single hydroxyl group results in naloxone, a strong μ-receptor antagonist. The structures of some of these compounds are shown later in this chapter. Some opioids, eg, nalbuphine, a mixed agonist-antagonist, are capable of producing an agonist (or partial agonist) effect at one opioid receptor subtype and an antagonist effect at another. The receptor-activating properties and affinities of opioid analgesics can be manipulated by pharmaceutical chemistry; in addition, certain opioid analgesics are modified in the liver, resulting in compounds with greater analgesic action. Chemically, the opioids derived from opium are phenanthrene derivatives and include four or more fused rings, while most of the synthetic opioids are simpler molecules. TABLE 31–1 Opioid receptor subtypes, their functions, and their endogenous peptide affinities.


Endogenous Opioid Peptides Opioid alkaloids (eg, morphine) produce analgesia through actions at central nervous system (CNS) receptors that also respond to certain endogenous peptides with opioid-like pharmacologic properties. The general term currently used for these endogenous substances is endogenous opioid peptides. Three families of endogenous opioid peptides have been described: the endorphins, the pentapeptide enkephalins (methionineenkephalin [met-enkephalin] and leucine-enkephalin [leu-enkephalin]), and the dynorphins. These three families of endogenous opioid peptides have overlapping affinities for opioid receptors (Table 31–1). The endogenous opioid peptides are derived from three precursor proteins: prepro-opiomelanocortin (POMC), preproenkephalin (proenkephalin A), and preprodynorphin (proenkephalin B). POMC contains the met-enkephalin sequence, β-endorphin, and several nonopioid peptides, including adrenocorticotropic hormone (ACTH), β-lipotropin, and melanocyte-stimulating hormone. Preproenkephalin contains six copies of met-enkephalin and one copy of leu-enkephalin. Leu- and met-enkephalin have slightly higher affinity for the δ (delta) than for the μ-opioid receptor (Table 31–1). Preprodynorphin yields several active opioid peptides that contain the leu-enkephalin sequence. These are dynorphin A, dynorphin B, and α and β neoendorphins. Painful stimuli can evoke release of endogenous opioid peptides under the stress associated with pain or the anticipation of pain, and they diminish the perception of pain. In contrast to the analgesic role of leu- and met-enkephalin, an analgesic action of dynorphin A—through its binding to κ-opioid receptors—remains controversial. Dynorphin A is also found in the dorsal horn of the spinal cord. Increased levels of dynorphin occur in the dorsal horn after tissue injury and inflammation. This elevated dynorphin level is proposed to increase pain and induce a state of longlasting sensitization and hyperalgesia. The pronociceptive action of dynorphin in the spinal cord appears to be independent of the opioid receptor system. This pronociceptive effect may involve an action via dynorphin A binding to the N-methyl-D-aspartate (NMDA)receptor complex and possibly to a novel receptor-ligand system homologous to the opioid peptides. The principal receptor for this novel system is the G protein-coupled orphanin opioid-receptor-like subtype 1 (ORL1). Its endogenous ligand has been termed nociceptin by one group of investigators and orphanin FQ by another group. This ligand-receptor system is currently known as the N/OFQ system. Nociceptin is structurally similar to dynorphin except for the absence of an N-terminal tyrosine; it acts only at the ORL1 receptor, now known as NOP. The N/OFQ system is widely expressed in the CNS and periphery,


reflecting its equally diverse biology and pharmacology. As a result of experiments using highly selective NOP receptor ligands, the N/OFQ system has been implicated in both pro- and anti-nociceptive activity as well as in the modulation of drug reward, learning, mood, anxiety, and cough processes, and of parkinsonism.

Pharmacokinetics Properties of clinically important opioids are summarized in Table 31–2. TABLE 31–2 Common opioid analgesics.



A. Absorption Most opioid analgesics are well absorbed when given by subcutaneous, intramuscular, and oral routes. However, because of the firstpass effect, the oral dose of the opioid (eg, morphine) to elicit a therapeutic effect may need to be much higher than the parenteral dose. As there is considerable interpatient variability in first-pass opioid metabolism, prediction of an effective oral dose is difficult. Certain analgesics such as codeine and oxycodone are effective orally because they have reduced first-pass metabolism. By avoiding first-pass metabolism, nasal insufflation of certain opioids can rapidly result in therapeutic blood levels. Other routes of opioid administration include oral mucosa via lozenges, and the transdermal route via patches. The latter can provide delivery of potent analgesics over days. B. Distribution The uptake of opioids by various organs and tissues is a function of both physiologic and chemical factors. Although all opioids bind to plasma proteins with varying affinity, the drugs rapidly leave the blood compartment and localize in highest concentrations in highly perfused tissues such as the brain, lungs, liver, kidneys, and spleen. Drug concentrations in skeletal muscle may be much lower, but this tissue serves as the main reservoir because of its greater bulk. Even though blood flow to fatty tissue is much lower than to the highly perfused tissues, accumulation can be very important, particularly after frequent high-dose administration or continuous infusion of highly lipophilic opioids that are slowly metabolized, eg, fentanyl. C. Metabolism The opioids are converted in large part to polar metabolites (mostly glucuronides), which are then readily excreted by the kidneys. For example, morphine, which contains free hydroxyl groups, is primarily conjugated to morphine-3-glucuronide (M3G), a compound with neuroexcitatory properties. The neuroexcitatory effects of M3G do not appear to be mediated by Ο receptors and are under further study. In contrast, approximately 10% of morphine is metabolized to morphine- 6-glucuronide (M6G), an active metabolite with analgesic potency four to six times that of its parent compound. However, these relatively polar metabolites have limited ability to cross the bloodbrain barrier and probably do not contribute significantly to the usual CNS effects of a single dose of morphine. Importantly, accumulation of these metabolites may produce unexpected adverse effects in patients with renal failure or when exceptionally large doses of morphine are administered or high doses are administered over long periods. This can result in M3G-induced CNS excitation (seizures) or enhanced and prolonged opioid action produced by M6G. CNS uptake of M3G and, to a lesser extent, M6G can be enhanced by coadministration of probenecid or of drugs that inhibit the P-glycoprotein drug transporter. 1. Hepatic P450 metabolism—Hepatic oxidative metabolism is the primary route of degradation of the phenylpiperidine opioids (fentanyl, meperidine, alfentanil, sufentanil) and eventually leaves only small quantities of the parent compound unchanged for excretion. However, accumulation of a demethylated metabolite of meperidine, normeperidine, may occur in patients with decreased renal function and in those receiving multiple high doses of the drug. In high concentrations, normeperidine may cause seizures. In contrast, no active metabolites of fentanyl have been reported. The P450 isozyme CYP3A4 metabolizes fentanyl by N-dealkylation in the liver. CYP3A4 is also present in the mucosa of the small intestine and contributes to the first-pass metabolism of fentanyl when it is taken orally. Codeine, oxycodone, and hydrocodone undergo metabolism in the liver by P450 isozyme CYP2D6, resulting in the production of metabolites of greater potency. For example, codeine is demethylated to morphine, which is then conjugated. Hydrocodone is metabolized to hydromorphone and, like morphine, hydromorphone is conjugated, yielding hydromorphone-3-glucuronide (H3G), which has CNS excitatory properties. Hydromorphone cannot form a 6-glucuronide metabolite. Similarly, oxycodone is metabolized to oxymorphone, which is then conjugated to oxymorphone-3-glucuronide (O3G). Genetic polymorphism of CYP2D6 has been documented and linked to the variation in analgesic and adverse responses seen among patients. In contrast, the metabolites of oxycodone and hydrocodone may be of minor consequence; the parent compounds are currently believed to be directly responsible for the majority of their analgesic actions. However, oxycodone and its metabolites can accumulate under conditions of renal failure and have been associated with prolonged action and sedation. In the case of codeine, conversion to morphine may be of greater importance because codeine itself has relatively low affinity for opioid receptors. As a result, some patients (so-called poor metabolizers) may experience no significant analgesic effect. In contrast, there have been case reports of an exaggerated response to codeine due to enhanced metabolic conversion to morphine (ie, ultra rapid metabolizers; see Chapters 4, 5) resulting in respiratory depression and death. For this reason, routine use of codeine, especially in pediatric age groups, is now being eliminated in the United States. 2. Plasma esterase metabolism—Esters (eg, heroin, remifentanil) are rapidly hydrolyzed by common plasma and tissue esterases. Heroin (diacetylmorphine) is hydrolyzed to monoacetylmorphine and finally to morphine, which is then conjugated with glucuronic acid. D. Excretion Polar metabolites, including glucuronide conjugates of opioid analgesics, are excreted mainly in the urine. Small amounts of unchanged drug may also be found in the urine. In addition, glucuronide conjugates are found in the bile, but enterohepatic circulation represents only a small portion of the excretory process of these polar metabolites. In patients with renal impairment the effects of active polar


metabolites should be considered before the administration of potent opioids such as morphine or hydromorphone—especially when given at high doses—due to the risk of sedation and respiratory depression.

Pharmacodynamics A. Mechanism of Action Opioid agonists produce analgesia by binding to specific G protein-coupled receptors that are located in brain and spinal cord regions involved in the transmission and modulation of pain (Figure 31–1). Some effects may be mediated by opioid receptors on peripheral sensory nerve endings.



FIGURE 31–1 Potential receptor mechanisms of analgesic drugs. The primary afferent neuron (cell body not shown) originates in the periphery and carries pain signals to the dorsal horn of the spinal cord, where it synapses via glutamate and neuropeptide transmitters with the secondary neuron. Pain stimuli can be attenuated in the periphery (under inflammatory conditions) by opioids acting at μ-opioid receptors (MOR) or blocked in the afferent axon by local anesthetics (not shown). Action potentials reaching the dorsal horn can be attenuated at the presynaptic ending by opioids and by calcium blockers (ziconotide), α2 agonists, and possibly, by drugs that increase synaptic concentrations of norepinephrine by blocking reuptake (tapentadol). Opioids also inhibit the postsynaptic neuron, as do certain neuropeptide antagonists acting at tachykinin (NK1) and other neuropeptide receptors. 1. Receptor types—As noted previously, three major classes of opioid receptors (μ, δ, and κ) have been identified in various nervous system sites and in other tissues (Table 31–1). Each of the three major receptors has now been cloned. All are members of the G protein-coupled family of receptors and show significant amino acid sequence homologies. Multiple receptor subtypes have been proposed based on pharmacologic criteria, including μ1 , μ2 ; δ1 , δ2 ; and κ1 , κ2 , and κ3 . However, genes encoding only one subtype from each of the μ, δ, and κ receptor families have thus far been isolated and characterized. One plausible explanation is that μ-receptor subtypes arise from alternate splice variants of a common gene. This idea has been supported by the identification of receptor splice variants in mice and humans, and a recent report pointed to the selective association of a μ-opioid receptor splice variant (MOR1D) with the induction of itch rather than the suppression of pain. Since an opioid may function with different potencies as an agonist, partial agonist, or antagonist at more than one receptor class or subtype, it is not surprising that these agents are capable of diverse pharmacologic effects. 2. Cellular actions—At the molecular level, opioid receptors form a family of proteins that physically couple to G proteins and through this interaction affect ion channel gating, modulate intracellular Ca2+ disposition, and alter protein phosphorylation (see Chapter 2). The opioids have two well-established direct Gi/0 protein-coupled actions on neurons: (1) they close voltage-gated Ca2+ channels on presynaptic nerve terminals and thereby reduce transmitter release, and (2) they open K+ channels and hyperpolarize and thus inhibit postsynaptic neurons. Figure 31–1 schematically illustrates these effects. The presynaptic action—depressed transmitter release—has been demonstrated for a large number of neurotransmitters, including glutamate, the principal excitatory amino acid released from nociceptive nerve terminals, as well as acetylcholine, norepinephrine, serotonin, and substance P. 3. Relation of physiologic effects to receptor type—The majority of currently available opioid analgesics act primarily at the μ-opioid receptor (Table 31–2). Analgesia and the euphoriant, respiratory depressant, and physical dependence properties of morphine result principally from actions at μ receptors. In fact, the μ receptor was originally defined using the relative potencies for clinical analgesia of a series of opioid alkaloids. However, opioid analgesic effects are complex and include interaction with δ and κ receptors. This is supported in part by the study of genetic knockouts of the μ, δ, and κ genes in mice. The development of μ-receptor–selective agonists could be clinically useful if their side-effect profiles (respiratory depression, risk of dependence) were more favorable than those found with current μ-receptor agonists, such as morphine. Although morphine does act at κ and δ receptor sites, it is unclear to what extent this contributes to its analgesic action. The endogenous opioid peptides differ from most of the alkaloids in their affinity for the δ and κ receptors (Table 31–1). In an effort to develop opioid analgesics with a reduced incidence of respiratory depression or propensity for addiction and dependence, compounds that show preference for κ opioid receptors have been developed. Butorphanol and nalbuphine have shown some clinical success as analgesics, but they can cause dysphoric reactions and have limited potency. It is interesting that butorphanol has also been shown to cause significantly greater analgesia in women than in men. In fact, gender-based differences in analgesia mediated by μ- and δ-receptor activation have been widely reported. 4. Receptor distribution and neural mechanisms of analgesia—Opioid receptor binding sites have been localized autoradiographically with high-affinity radioligands and with antibodies to unique peptide sequences in each receptor subtype. All three major receptors are present in high concentrations in the dorsal horn of the spinal cord. Receptors are present both on spinal cord pain transmission neurons and on the primary afferents that relay the pain message to them (Figure 31–2, sites A and B). Although opioid agonists directly inhibit dorsal horn pain transmission neurons, they also inhibit the release of excitatory transmitters from the primary afferents. Although there are reports that heterodimerization of the μ-opioid and δ-opioid receptors contributes to μ-agonist efficacy (eg, inhibition of presynaptic voltage-gated calcium channel activity), a recent study using a transgenic mouse that expresses a δ-receptor–enhanced green fluorescent protein (eGFP) fusion protein showed little overlap of μ receptor and δ receptor in dorsal root ganglion neurons. Importantly, the μ receptor is associated with TRPV1 and peptide (substance P)-expressing nociceptors, whereas δ-receptor expression predominates in the non-peptidergic population of nociceptors, including many primary afferents with myelinated axons. This finding is consistent with the action of intrathecal μ-receptor– and δ-receptor–selective ligands that are found to block heat versus mechanical pain processing, respectively. Very recently, an association of the δ but not the μ receptor with large diameter mechanoreceptive afferents has been described. To what extent the differential expression of the μ receptor and δ receptor in the dorsal root ganglia is characteristic of neurons throughout the CNS remains to be determined.


FIGURE 31–2 Putative sites of action of opioid analgesics. Sites of action on the afferent pain transmission pathway from the periphery to the higher centers are shown. A: Direct action of opioids on inflamed or damaged peripheral tissues (see Figure 31–1 for detail). B: Inhibition also occurs in the spinal cord (see Figure 31–1). C: Possible sites of action in the thalamus. The fact that opioids exert a powerful analgesic effect directly on the spinal cord has been exploited clinically by direct application of opioid agonists to the spinal cord. This spinal action provides a regional analgesic effect while reducing the unwanted respiratory depression, nausea and vomiting, and sedation that may occur from the supraspinal actions of systemically administered opioids. Under most circumstances, opioids are given systemically and thus act simultaneously at multiple sites. These include not only the ascending pathways of pain transmission beginning with specialized peripheral sensory terminals that transduce painful stimuli (Figure 31–2) but also descending (modulatory) pathways (Figure 31–3). At these sites as at others, opioids directly inhibit neurons; yet this action results in the activation of descending inhibitory neurons that send processes to the spinal cord and inhibit pain transmission neurons. This activation has been shown to result from the inhibition of inhibitory neurons in several locations (Figure 31–4). Taken together, interactions at these sites increase the overall analgesic effect of opioid agonists.


FIGURE 31–3 Brainstem local circuitry underlying the modulating effect of μ-opioid receptor (MOR)–mediated analgesia on descending pathways. The pain-inhibitory neuron is indirectly activated by opioids (exogenous or endogenous), which inhibit an inhibitory (GABAergic) interneuron. This results in enhanced inhibition of nociceptive processing in the dorsal horn of the spinal cord (see Figure 31–4).


FIGURE 31–4 Opioid analgesic action on the descending inhibitory pathway. Sites of action of opioids on pain-modulating neurons in the midbrain and medulla including the midbrain periaqueductal gray area (A), rostral ventral medulla (B), and the locus caeruleus indirectly control pain transmission pathways by enhancing descending inhibition to the dorsal horn (C). When pain-relieving opioid drugs are given systemically, they presumably act upon neuronal circuits normally regulated by endogenous opioid peptides and part of the pain-relieving action of exogenous opioids may involve the release of endogenous opioid peptides. For example, an exogenous opioid agonist (eg, morphine) may act primarily and directly at the μ receptor, but this action may evoke the release of endogenous opioids that additionally act at δ and κ receptors. Thus, even a receptor-selective ligand can initiate a complex sequence of events involving multiple synapses, transmitters, and receptor types. Animal and human clinical studies demonstrate that both endogenous and exogenous opioids can also produce analgesia at sites outside the CNS. Pain associated with inflammation seems especially sensitive to these peripheral opioid actions. The presence of functional μ receptors on the peripheral terminals of sensory neurons supports this hypothesis. Furthermore, activation of peripheral μ receptors results in a decrease in sensory neuron activity and transmitter release. The endogenous release of β-endorphin produced by immune cells within injured or inflamed tissue represents one source of physiologic peripheral μ-receptor activation. Intra-articular administration of opioids, eg, following arthroscopic knee surgery, has shown clinical benefit for up to 24 hours. For this reason opioids selective for a peripheral site of action may be useful adjuncts in the treatment of inflammatory pain (see Box: Ion Channels & Novel Analgesic Targets). Such compounds could have the additional benefit of reducing unwanted effects such as nausea. 5. Tolerance and dependence—With frequently repeated therapeutic doses of morphine or its surrogates, there is a gradual loss in effectiveness; this loss of effectiveness is termed tolerance. To reproduce the original response, a larger dose must be administered. Along with tolerance, physical dependence develops. Physical dependence is defined as a characteristic withdrawal or abstinence


syndrome when a drug is stopped or an antagonist is administered (see also Chapter 32). The mechanism of development of opioid tolerance and physical dependence is poorly understood, but persistent activation of μ receptors such as occurs with the treatment of severe chronic pain appears to play a primary role in its induction and maintenance. Current concepts have shifted away from tolerance being driven by a simple up-regulation of the cyclic adenosine monophosphate (cAMP) system. Although this process is associated with tolerance, it is not sufficient to explain it. A second hypothesis for the development of opioid tolerance and dependence is based on the concept of receptor recycling. Normally, activation of μ receptors by endogenous ligands results in receptor endocytosis followed by resensitization and recycling of the receptor to the plasma membrane (see Chapter 2). However, using genetically modified mice, research now shows that the failure of morphine to induce endocytosis of the μopioid receptor is an important component of tolerance and dependence. In further support of this idea, methadone, a μ-receptor agonist used for the treatment of opioid tolerance and dependence, induces receptor endocytosis. This suggests that maintenance of normal sensitivity of μ receptors requires reactivation by endocytosis and recycling. The concept of receptor uncoupling has also gained prominence. Under this hypothesis, tolerance results from a dysfunction of structural interactions between the μ receptor and G proteins, second-messenger systems, and their target ion channels. Uncoupling and recoupling of μ receptor function is likely linked to receptor recycling. Moreover, the NMDA-receptor ion channel complex has been shown to play a critical role in tolerance development and maintenance. Consistent with this hypothesis, NMDA-receptor antagonists such as ketamine can block tolerance development. Although a role in endocytosis is not yet clearly defined, the development of novel NMDA-receptor antagonists or other strategies to recouple μ receptors to their target ion channels provides hope for achieving a clinically effective means to prevent or reverse opioid analgesic tolerance. 6. Opioid-induced hyperalgesia—In addition to the development of tolerance, persistent administration of opioid analgesics can increase the sensation of pain, resulting in a state of hyperalgesia. This phenomenon can be produced with several opioid analgesics, including morphine, fentanyl, and remifentanil. Spinal dynorphin and activation of the bradykinin and NMDA receptors have emerged as important candidates for the mediation of opioid-induced hyperalgesia. This is one more reason why the use of opioids for chronic pain is controversial. B. Organ System Effects of Morphine and Its Surrogates The actions described below for morphine, the prototypic opioid agonist, can also be observed with other opioid agonists, partial agonists, and those with mixed receptor effects. Characteristics of specific members of these groups are discussed below. 1. Central nervous system effects—The principal effects of opioid analgesics with affinity for μ receptors are on the CNS; the more important ones include analgesia, euphoria, sedation, and respiratory depression. With repeated use, a high degree of tolerance occurs to all of these effects (Table 31–3). TABLE 31–3 Degrees of tolerance that may develop to some of the effects of the opioids.


Ion Channels & Novel Analgesic Targets Even the most severe acute pain (lasting hours to days) can usually be well controlled—with significant but tolerable adverse effects—using currently available analgesics, especially the opioids. Chronic pain (lasting weeks to months), however, is not very satisfactorily managed with opioids. It is now known that in chronic pain, receptors on sensory nerve terminals in the periphery contribute to increased excitability of sensory nerve endings (peripheral sensitization). The hyperexcitable sensory neuron bombards the spinal cord, leading to increased excitability and synaptic alterations in the dorsal horn (central sensitization). Such changes are likely important contributors to chronic inflammatory and neuropathic pain states. In the effort to discover better analgesic drugs for chronic pain, renewed attention is being paid to the molecular basis of peripheral sensory transduction. Potentially important ion channels associated with the primary afferent nociceptor include members of the transient receptor potential family, notably the capsaicin receptor, TRPV1 (which is activated by multiple noxious stimuli such as heat, protons, and products of inflammation) as well as TRPA1, activated by inflammatory mediators; and P2X receptors (which are responsive to purines released from tissue damage). Special subtypes of voltage-gated sodium channels (Nav 1.7, 1.8, 1.9) are uniquely associated with nociceptive neurons in dorsal root ganglia. Lidocaine and mexiletine, which are useful in some chronic pain states, may act by blocking this class of channels. Certain centipede toxins appear to selectively inhibit Nav 1.7 channels and may also be useful in the treatment of chronic pain. Genetic polymorphisms of Nav 1.7 are associated with either absence or predisposition to pain. Because of the importance of their peripheral sites of action, therapeutic strategies that deliver agents that block peripheral pain transduction or transmission have been introduced in the form of transdermal patches and balms. In addition, products that systemically target peripheral TRPV1, TRPA1 and sodium channel function are in development. Ziconotide, a blocker of voltage-gated N-type calcium channels, is approved for intrathecal analgesia in patients with refractory chronic pain. Ziconotide is a synthetic peptide related to the marine snail toxin ω-conotoxin, which selectively blocks Ntype calcium channels. Gabapentin/pregabalin, anticonvulsant analogs of GABA (see Chapter 24) that are effective treatments for neuropathic (nerve injury) pain act at the α2δ1 subunit of voltage-gated calcium channels. N-methyl-d-aspartate (NMDA) receptors appear to play a very important role in central sensitization at both spinal and supraspinal levels. Although certain NMDA antagonists have demonstrated analgesic activity (eg, ketamine), it has been difficult to find agents with an acceptably low profile of adverse effects or neurotoxicity. However, ketamine infused at very small doses improves analgesia and can reduce opioid requirements under conditions of opioid tolerance, eg, after major abdominal and spinal surgery. GABA and acetylcholine (through nicotinic receptors) appear to control the central synaptic release of several transmitters involved in nociception. Nicotine itself and certain nicotine analogs cause analgesia, and their use for postoperative analgesia is under investigation. Use of antibodies that bind nerve growth factor (NGF) has been shown to block inflammatory and back pain and is awaiting FDA approval. Finally, work on cannabinoids and vanilloids and their receptors suggest that Δ9- tetrahydrocannabinol, which acts primarily on CB1 cannabinoid receptors, can synergize with μ-receptor analgesics and interact with the TRPV1 capsaicin receptor to produce analgesia under certain circumstances. As our understanding of peripheral and central pain transduction improves, additional therapeutic targets and strategies will become available. Combined with our present knowledge of opioid analgesics, a “multimodal” approach to pain therapy is emerging. Multimodal analgesia involves the administration of multiple agents (eg, NSAIDs, gabapentinoids, selective norepinephrine receptor inhibitors, etc) with complementary mechanisms of action to provide analgesia that is superior to that provided by an individual compound. Another benefit of multimodal analgesia is reduced opioid requirements with fewer adverse effects. a. Analgesia—Pain consists of both sensory and affective (emotional) components. Opioid analgesics are unique in that they can reduce both aspects of the pain experience. In contrast, nonsteroidal anti-inflammatory analgesic drugs, eg, ibuprofen, have no significant effect on the emotional aspects of pain. b. Euphoria—Typically, patients or intravenous drug users who receive intravenous morphine experience a pleasant floating sensation with lessened anxiety and distress. However, dysphoria, an unpleasant state characterized by restlessness and malaise, may also occur. c. Sedation—Drowsiness and clouding of mentation are common effects of opioids. There is little or no amnesia. Sleep is induced by opioids more frequently in the elderly than in young, healthy individuals. Ordinarily, the patient can be easily aroused from this sleep. However, the combination of morphine with other central depressant drugs such as the sedative-hypnotics may result in very deep sleep. Marked sedation occurs more frequently with compounds closely related to the phenanthrene derivatives and less frequently with the synthetic agents such as meperidine and fentanyl. In standard analgesic doses, morphine (a phenanthrene) disrupts normal rapid eye movement (REM) and non-REM sleep patterns. This disrupting effect is probably characteristic of all opioids. In contrast to humans, a number of other species (cats, horses, cows, pigs) may manifest excitation rather than sedation when given opioids. These paradoxical effects are at least partially dose-dependent. d. Respiratory depression—All of the opioid analgesics can produce significant respiratory depression by inhibiting brainstem


respiratory mechanisms. Alveolar P CO2 may increase, but the most reliable indicator of this depression is a depressed response to a carbon dioxide challenge. The respiratory depression is dose-related and is influenced significantly by the degree of sensory input occurring at the time. For example, it is possible to partially overcome opioid-induced respiratory depression by a variety of stimuli. When strongly painful stimuli that have prevented the depressant action of a large dose of an opioid are relieved, respiratory depression may suddenly become marked. A small to moderate decrease in respiratory function, as measured by Pa CO2 elevation, may be well tolerated in the patient without prior respiratory impairment. However, in individuals with increased intracranial pressure, asthma, chronic obstructive pulmonary disease, or cor pulmonale, this decrease in respiratory function may not be tolerated. Opioid-induced respiratory depression remains one of the most difficult clinical challenges in the treatment of severe pain. Ongoing research to overcome this problem is focused on μ-receptor pharmacology and serotonin signaling pathways in the brainstem respiratory control centers. e. Cough suppression—Suppression of the cough reflex is a well-recognized action of opioids. Codeine in particular has been used to advantage in persons suffering from pathologic cough. However, cough suppression by opioids may allow accumulation of secretions and thus lead to airway obstruction and atelectasis. f. Miosis—Constriction of the pupils is seen with virtually all opioid agonists. Miosis is a pharmacologic action to which little or no tolerance develops, even in highly tolerant addicts (Table 31–3); thus, it is valuable in the diagnosis of opioid overdose. This action, which can be blocked by opioid antagonists, is mediated by parasympathetic pathways, which, in turn, can be blocked by atropine. g. Truncal rigidity—Several opioids can intensify tone in the large trunk muscles. It was originally believed that truncal rigidity involved a spinal cord action of these drugs, but a supraspinal action is likely. Truncal rigidity reduces thoracic compliance and thus interferes with ventilation. The effect is most apparent when high doses of the highly lipid-soluble opioids (eg, fentanyl, sufentanil, alfentanil, remifentanil) are rapidly administered intravenously. Truncal rigidity may be overcome by administration of an opioid antagonist, which of course will also antagonize the analgesic action of the opioid. Preventing truncal rigidity while preserving analgesia requires the concomitant use of neuromuscular blocking agents. h. Nausea and vomiting—The opioid analgesics can activate the brainstem chemoreceptor trigger zone to produce nausea and vomiting. As ambulation seems to increase the incidence of nausea and vomiting there may also be a vestibular component in this effect. i. Temperature—Homeostatic regulation of body temperature is mediated in part by the action of endogenous opioid peptides in the brain. For example, administration of μ-opioid receptor agonists, such as morphine to the anterior hypothalamus produces hyperthermia, whereas administration of κ agonists induces hypothermia. j. Sleep architecture—Although the mechanism by which opioids interact with circadian rhythm is unclear, they can decrease the percentage of stage 3 and 4 sleep, which may result in fatigue and other sleep disorders, including sleep-disordered breathing and central sleep apnea. 2. Peripheral effects a. Cardiovascular system—Most opioids have no significant direct effects on the heart and, other than bradycardia, no major effects on cardiac rhythm. Meperidine is an exception to this generalization because its antimuscarinic action can result in tachycardia. Blood pressure is usually well maintained in subjects receiving opioids unless the cardiovascular system is stressed, in which case hypotension may occur. This hypotensive effect is probably due to peripheral arterial and venous dilation, which has been attributed to a number of mechanisms including central depression of vasomotor-stabilizing mechanisms and release of histamine. No consistent effect on cardiac output is seen, and the electrocardiogram is not significantly affected. However, caution should be exercised in patients with decreased blood volume, because the above mechanisms make these patients susceptible to hypotension. Opioid analgesics affect cerebral circulation minimally except when P CO2 rises as a consequence of respiratory depression. Increased P CO2 leads to cerebral vasodilation associated with a decrease in cerebral vascular resistance, an increase in cerebral blood flow, and an increase in intracranial pressure. b. Gastrointestinal tract—Constipation has long been recognized as an effect of opioids, an effect that does not diminish with continued use. That is, tolerance does not develop to opioid-induced constipation (Table 31–3). Opioid receptors exist in high density in the gastrointestinal tract, and the constipating effects of the opioids are mediated through an action on the enteric nervous system (see Chapter 6) as well as the CNS. In the stomach, motility (rhythmic contraction and relaxation) may decrease but tone (persistent contraction) may increase—particularly in the central portion; gastric secretion of hydrochloric acid is decreased. Small intestine resting tone is increased, with periodic spasms, but the amplitude of nonpropulsive contractions is markedly decreased. In the large intestine, propulsive peristaltic waves are diminished and tone is increased; this delays passage of the fecal mass and allows increased absorption of water, which leads to constipation. The large bowel actions are the basis for the use of opioids in the management of diarrhea, and constipation is a major problem in the use of opioids for control of severe cancer pain. c. Biliary tract—The opioids contract biliary smooth muscle, which can result in biliary colic. The sphincter of Oddi may constrict,


resulting in reflux of biliary and pancreatic secretions and elevated plasma amylase and lipase levels. d. Renal—Renal function is depressed by opioids. It is believed that in humans this is chiefly due to decreased renal plasma flow. In addition, μ opioids have an antidiuretic effect in humans. Mechanisms may involve both the CNS and peripheral sites. Opioids also enhance renal tubular sodium reabsorption. The role of opioid-induced changes in antidiuretic hormone (ADH) release is controversial. Ureteral and bladder tone are increased by therapeutic doses of the opioid analgesics. Increased sphincter tone may precipitate urinary retention, especially in postoperative patients. Occasionally, ureteral colic caused by a renal calculus is made worse by opioid-induced increase in ureteral tone. e. Uterus—The opioid analgesics may prolong labor. Although the mechanism for this action is unclear, both μ- and κ-opioid receptors are expressed in human uterine muscle. Fentanyl and meperidine (pethidine) inhibit uterine contractility but only at supraclinical concentrations; morphine had no reported effects. In contrast, the κ agonist [3H]-D-ala2,L-met5-enkephalinamide (DAMEA) inhibits contractility in human uterine muscle strips. f. Endocrine—Opioids stimulate the release of ADH, prolactin, and somatotropin but inhibit the release of luteinizing hormone (Table 31–1). These effects suggest that endogenous opioid peptides, through effects in the hypothalamus, modulate these systems. Patients receiving chronic opioid therapy can have low testosterone resulting in decreased libido, energy, and mood. Women can experience dysmenorrhea or amenorrhea. g. Pruritus—The opiates, such as morphine and codeine, produce flushing and warming of the skin accompanied sometimes by sweating, urticaria, and itching. Although peripheral histamine release is an important contributor, all opioids can cause pruritus via a central (spinal cord and medullary) action on pruritoceptive neural circuits. When opioids are administered to the neuraxis by the spinal or epidural route, their usefulness may be limited by intense pruritus over the lips and torso. The incidence of opioid-induced pruritus via the neuraxial route is high, estimated at 70–100%. However, studies have demonstrated the efficacy of selective κ agonists (eg, nalfurafine) in the treatment of itch. h. Immune—The opioids modulate the immune system by effects on lymphocyte proliferation, antibody production, and chemotaxis. In addition, leucocytes migrate to the site of tissue injury and release opioid peptides, which in turn help counter inflammatory pain. However, natural killer cell cytolytic activity and lymphocyte proliferative responses to mitogens are usually inhibited by opioids, which may play a role in tumor progression. Although the mechanisms involved are complex, activation of central opioid receptors could mediate a significant component of the changes observed in peripheral immune function. These effects are mediated by the sympathetic nervous system in the case of acute administration and by the hypothalamic-pituitary-adrenal system in the case of prolonged administration of opioids.

CLINICAL PHARMACOLOGY OF THE OPIOID ANALGESICS Successful treatment of pain is a challenging task that begins with careful attempts to assess the source and magnitude of the pain. The amount of pain experienced by the patient is often measured by means of a pain Numeric Rating Scale (NRS) or less frequently by marking a line on a Visual Analog Scale (VAS) with word descriptors ranging from no pain (0) to excruciating pain (10). In either case, values indicate the magnitude of pain as: mild (1–3), moderate (4–6), or severe (7–10). A similar scale can be used with children (Face, Legs, Activity, Cry, Consolability [FLACC] or Wong-Baker scales) and with patients who cannot speak; the Wong-Baker scale depicts five faces ranging from smiling (no pain) to crying (maximum pain). There are specialized scales for patients with specific conditions including rheumatoid arthritis and dementia. More comprehensive questionnaires such as the McGill Pain Questionnaire address the multiple facets of pain. For a patient in severe pain, administration of an opioid analgesic is usually considered a primary part of the overall management plan. Determining the route of administration (oral, parenteral, neuraxial), duration of drug action, ceiling effect (maximal intrinsic activity), duration of therapy, potential for adverse effects, and the patient’s past experience with opioids all should be addressed. One of the principal errors made by physicians in this setting is failure to assess adequately a patient’s pain and to match its severity with an appropriate level of therapy. Just as important is the principle that following delivery of the therapeutic plan, its effectiveness must be reevaluated and the plan modified, if necessary, if the response was excessive or inadequate. Use of opioid drugs in acute situations should be contrasted with their use in chronic pain management, in which a multitude of other factors must be considered, including the development of tolerance to and physical dependence on opioid analgesics.

Clinical Use of Opioid Analgesics A. Analgesia Severe, constant pain is usually relieved with opioid analgesics having high intrinsic activity (see Table 31–2), whereas sharp, intermittent pain does not appear to be as effectively controlled.


The pain associated with cancer and other terminal illnesses must be treated aggressively and often requires a multidisciplinary approach for effective management. Such conditions may require continuous use of potent opioid analgesics and are associated with some degree of tolerance and dependence. However, this should not be used as a barrier to providing patients with the best possible care and quality of life. The World Health Organization Ladder (see http://www.who.int/cancer/palliative/painladder/en/) was created in 1986 to promote awareness of the optimal treatment of pain for individuals with cancer and has helped improve pain care for cancer patients worldwide. Research in the hospice setting has also demonstrated that fixed-interval administration of opioid medication (ie, a regular dose at a scheduled time) is more effective in achieving pain relief than dosing on demand. New dosage forms of opioids that allow slower release of the drug are now available, eg, sustained-release forms of morphine (MS Contin) and oxycodone (OxyContin). Their purported advantage is a longer and more stable level of analgesia. However, there is little evidence to support longterm (greater than 6 months) use of sustained release opioids to manage chronic pain in the non-cancer patient. If disturbances of gastrointestinal function prevent the use of oral sustained-release morphine, then a fentanyl transdermal system (fentanyl patch) can be used over long periods. Furthermore, buccal transmucosal fentanyl can be used for short episodes of breakthrough pain (see Alternative Routes of Administration). Administration of strong opioids by nasal insufflation is also efficacious, and nasal preparations are now available in some countries. Approval of such formulations in the USA is growing. In addition, stimulant drugs such as the amphetamines can enhance the analgesic actions of opioids and thus may be very useful adjuncts in the patient with chronic pain. Opioid analgesics are often used during obstetric labor. Because opioids cross the placental barrier and reach the fetus, care must be taken to minimize neonatal depression. If it occurs, immediate injection of the antagonist naloxone will reverse the depression. The phenylpiperidine drugs (eg, meperidine) appear to produce less depression, particularly respiratory depression, in newborn infants than does morphine; this may justify their use in obstetric practice. The acute, severe pain of renal and biliary colic often requires a strong agonist opioid for adequate relief. However, the drug-induced increase in smooth muscle tone may cause a paradoxical increase in pain secondary to increased spasm. An increase in the dose of opioid is usually successful in providing adequate analgesia. B. Acute Pulmonary Edema The relief produced by intravenous morphine in patients with dyspnea from pulmonary edema associated with left ventricular heart failure is remarkable. Proposed mechanisms include reduced anxiety (perception of shortness of breath) and reduced cardiac preload (reduced venous tone) and afterload (decreased peripheral resistance). However, if respiratory depression is a problem, furosemide may be preferred for the treatment of pulmonary edema. On the other hand, morphine can be particularly useful when treating painful myocardial ischemia with pulmonary edema. C. Cough Suppression of cough can be obtained at doses lower than those needed for analgesia. However, in recent years the use of opioid analgesics to allay cough has diminished largely because of the availability of a number of effective synthetic compounds that are neither analgesic nor addictive. These agents are discussed below. D. Diarrhea Diarrhea from almost any cause can be controlled with the opioid analgesics, but if diarrhea is associated with infection such use must not substitute for appropriate chemotherapy. Crude opium preparations (eg, paregoric) were used in the past to control diarrhea, but now synthetic surrogates with more selective gastrointestinal effects and few or no CNS effects, eg, diphenoxylate or loperamide, are used. Several preparations are available specifically for this purpose (see Chapter 62). E. Shivering Although all opioid agonists have some propensity to reduce shivering, meperidine is reported to have the most pronounced anti-shivering properties. Meperidine apparently blocks shivering mainly through an action on subtypes of the ι2 adrenoceptor. F. Applications in Anesthesia The opioids are frequently used as premedicant drugs before anesthesia and surgery because of their sedative, anxiolytic, and analgesic properties. They are also used intraoperatively both as adjuncts to other anesthetic agents and, in high doses (eg, 0.02–0.075 mg/kg of fentanyl), as a primary component of the anesthetic regimen (see Chapter 25). Opioids are most commonly used in cardiovascular surgery and other types of high-risk surgery in which a primary goal is to minimize cardiovascular depression. In such situations, mechanical respiratory assistance must be provided. Because of their direct action on the neurons of the superficial dorsal horn of the spine, opioids can also be used as regional analgesics, by administration into the epidural or subarachnoid spaces of the spinal column. A number of studies have demonstrated that long-lasting analgesia with minimal adverse effects can be achieved by epidural administration of 3–5 mg of morphine, followed by slow


infusion through a catheter placed in the epidural space. It was initially assumed that the epidural application of opioids might selectively produce analgesia without impairment of motor, autonomic, or sensory functions other than pain. However, respiratory depression can occur after the drug is injected into the epidural space and may require reversal with naloxone. Effects such as pruritus and nausea and vomiting are common after epidural and subarachnoid administration of opioids and may also be reversed with naloxone. Currently, the epidural route is favored over subarachnoid administration because adverse effects are less common and robust outcome studies have shown a significant reduction in perioperative mortality and morbidity with the use of thoracic epidural analgesia. The use of low doses of local anesthetics in combination with fentanyl infused through a thoracic epidural catheter has become an accepted method of pain control in patients recovering from thoracic and major upper abdominal surgery. In rare cases, chronic pain management specialists may elect to implant surgically a programmable infusion pump connected to a spinal catheter for continuous infusion of opioids or other analgesic compounds. G. Alternative Routes of Administration Patient-controlled analgesia (PCA) is widely used for the management of breakthrough pain. With PCA, the patient controls a parenteral (usually intravenous) infusion device by pressing a button to deliver a preprogrammed dose of the desired opioid analgesic. A programmable lockout interval prevents administration of another dose for a set period of time. Claims of better patient satisfaction are supported by well-designed clinical trials, making this approach very useful in postoperative pain control. However, health care personnel must be very familiar with the use of PCAs to avoid overdosage secondary to misuse or improper programming. There is a proven risk of PCA-associated respiratory depression and hypoxia that requires careful monitoring of vital signs and sedation level, and provision of supplemental oxygen. Continuous pulse oximetry is also recommended for patients receiving PCA-administered opioids; this is not a failsafe method for early detection of hypoventilation or apnea but rather serves as a safety net for an unrecognized adverse event. The risk of sedation is increased if medications with sedative properties, such as benzodiazepines and certain types of antiemetics, are concurrently prescribed. Rectal suppositories of morphine and hydromorphone have been used when oral and parenteral routes are undesirable. The transdermal fentanyl patch provides stable blood levels of drug and better pain control while avoiding the need for repeated parenteral injections. Fentanyl is the most successful opioid in transdermal application and is indicated for the management of persistent unremitting pain. Because of the complication of fentanyl-induced respiratory depression, the Food and Drug Administration (FDA) recommends that introduction of a transdermal fentanyl patch (25 mcg/h) be reserved for patients with an established oral morphine requirement of at least 60 mg/d for 1 week or more. Extreme caution must be exercised in any patient initiating therapy or undergoing a dose increase because the peak effects may not be realized until 24–48 hours after patch application. The buprenorphine patch (BuTrans) is an example of the transdermal delivery of a mixed agonist-antagonist for the treatment of chronic pain in addition to opioid maintenance or detoxification. The intranasal route avoids repeated parenteral drug injections and the first-pass metabolism of orally administered drugs. Butorphanol is the only opioid currently available in the USA in a nasal formulation, but more are expected. Another alternative to parenteral administration is the buccal transmucosal route, which uses a fentanyl citrate lozenge or a “lollipop” mounted on a stick.

Toxicity & Undesired Effects Direct toxic effects of the opioid analgesics that are extensions of their acute pharmacologic actions include respiratory depression, nausea, vomiting, and constipation (Table 31–4). Tolerance, dependence, diagnosis and treatment of overdosage, and contraindications must be considered. TABLE 31–4 Adverse effects of the opioid analgesics.


A. Tolerance and Dependence Drug dependence of the opioid type is marked by a relatively specific withdrawal or abstinence syndrome. Just as there are pharmacologic differences between the various opioids, there are also differences in psychological dependence and the severity of withdrawal effects. For example, withdrawal from dependence on a strong agonist is associated with more severe withdrawal signs and symptoms than withdrawal from a mild or moderate agonist. Administration of an opioid antagonist to an opioid-dependent person is followed by brief but severe withdrawal symptoms (see antagonist-precipitated withdrawal, below). The potential for physical and psychological dependence of the partial agonist-antagonist opioids appears to be less than that of the strong agonist drugs. 1. Opioid tolerance—is the phenomenon whereby repeated doses of opioids have a diminishing analgesic effect. Clinically, it has been described as an increasing opioid dose requirement to achieve the analgesia observed at the initiation of opioid administration. Although development of tolerance begins with the first dose of an opioid, tolerance may not become clinically manifest until after 2–3 weeks of frequent exposure to ordinary therapeutic doses. Nevertheless, perioperative and critical care use of ultrapotent opioid analgesics such as remifentanil have been shown to induce opioid tolerance within hours. Tolerance develops most readily when large doses are given at short intervals and is minimized by giving small amounts of drug with longer intervals between doses. A high degree of tolerance may develop to the analgesic, sedating, and respiratory depressant effects of opioid agonists. It is possible to produce respiratory arrest in a nontolerant person with a dose of 60 mg of morphine. However, in a patient who is opioid-dependent or requires escalating opioid administration to manage intractable cancer pain, doses such as 2000 mg of morphine taken over a 2- or 3-hour period may not produce significant respiratory depression. Tolerance also develops to the antidiuretic, emetic, and hypotensive effects but not to the miotic, convulsant, and constipating actions (Table 31–3). Following discontinuation of opioids, loss of tolerance to the sedating and respiratory effects of opioids is variable, and difficult to predict. However, tolerance to the emetic effects may persist for several months after withdrawal of the drug. Therefore, opioid tolerance differs by effect, drug, time, and the individual (genetic-epigenetic factors). Tolerance also develops to analgesics with mixed receptor effects but to a lesser extent than to the agonists. Adverse effects such as hallucinations, sedation, hypothermia, and respiratory depression are reduced after repeated administration of the mixed receptor drugs. However, tolerance to the latter agents does not generally include cross-tolerance to the agonist opioids. It is also important to note that tolerance does not develop to the antagonist actions of the mixed agents or to those of the pure antagonists. Cross-tolerance is an extremely important characteristic of the opioids, ie, patients tolerant to morphine often show a reduction in


analgesic response to other agonist opioids. This is particularly true of those agents with primarily μ-receptor agonist activity. Morphine and its congeners exhibit cross-tolerance not only with respect to their analgesic actions but also to their euphoriant, sedative, and respiratory effects. However, the cross-tolerance existing among the μ-receptor agonists can often be partial or incomplete. This clinical observation has led to the concept of “opioid rotation,” which has been used for many years in the treatment of cancer pain. A patient who is experiencing decreasing effectiveness of one opioid analgesic regimen is “rotated” to a different opioid analgesic (eg, morphine to hydromorphone; hydromorphone to methadone) and typically experiences significantly improved analgesia at a reduced overall equivalent dosage. Another approach is to recouple opioid receptor function as described previously through the use of adjunctive nonopioid agents. NMDA-receptor antagonists (eg, ketamine) have shown promise in preventing or reversing opioid-induced tolerance in animals and humans. Use of ketamine is increasing because well-controlled studies have shown clinical efficacy in reducing postoperative pain and opioid requirements in opioid-tolerant patients. Agents that independently enhance μ-receptor recycling may also hold promise for improving analgesia in the opioid-tolerant patient. 2. Dependence—The development of physical dependence is an invariable accompaniment of tolerance to repeated administration of an opioid of the μ type. Failure to continue administering the drug results in a characteristic withdrawal or abstinence syndrome that reflects an exaggerated rebound from the acute pharmacologic effects of the opioid. The signs and symptoms of withdrawal include rhinorrhea, lacrimation, yawning, chills, gooseflesh (piloerection), hyperventilation, hyperthermia, mydriasis, muscular aches, vomiting, diarrhea, anxiety, and hostility. The number and intensity of the signs and symptoms are largely dependent on the degree of physical dependence that has developed. Administration of an opioid at this time suppresses abstinence signs and symptoms almost immediately. The time of onset, intensity, and duration of abstinence syndrome depend on the drug previously used and may be related to its biologic half-life. With morphine or heroin, withdrawal signs usually start within 6–10 hours after the last dose. Peak effects are seen at 36–48 hours, after which most of the signs and symptoms gradually subside. By 5 days, most of the effects have disappeared, but some may persist for months. In the case of meperidine, the withdrawal syndrome largely subsides within 24 hours, whereas with methadone several days are required to reach the peak of the abstinence syndrome, and it may last as long as 2 weeks. The slower subsidence of methadone effects is associated with a less intense immediate syndrome, and this is the basis for its use in the detoxification of heroin addicts. However, despite the loss of physical dependence on the opioid, craving for it may persist. In addition to methadone, buprenorphine and the α2 agonist clonidine are FDA-approved treatments for opioid analgesic detoxification (see Chapter 32). A transient, explosive abstinence syndrome—antagonist-precipitated withdrawal—can be induced in a subject physically dependent on opioids by administering naloxone or another antagonist. Within 3 minutes after injection of the antagonist, signs and symptoms similar to those seen after abrupt discontinuance appear, peaking in 10–20 minutes and largely subsiding after 1 hour. Even in the case of methadone, withdrawal of which results in a relatively mild abstinence syndrome, the antagonist-precipitated abstinence syndrome may be very severe. In the case of agents with mixed effects, withdrawal signs and symptoms can be induced after repeated administration followed by abrupt discontinuance of pentazocine, cyclazocine, or nalorphine, but the syndrome appears to be somewhat different from that produced by morphine and other agonists. Anxiety, loss of appetite and body weight, tachycardia, chills, increase in body temperature, and abdominal cramps have been noted. 3. Addiction—As defined by the American Society of Addiction Medicine, addiction is a primary, chronic disease of brain reward, motivation, memory, and related circuitry. Dysfunction in these circuits leads to characteristic biologic, psychological, and social manifestations. This is reflected in an individual’s pathologic pursuit of reward and relief through substance use and other behaviors. Addiction is characterized by inability to abstain consistently, impairment in behavioral control, craving, diminished recognition of significant problems with one’s behaviors and interpersonal relationships, and a dysfunctional emotional response (see Chapter 32). The risk of inducing dependence and, potentially, addiction is clearly an important consideration in the therapeutic use of opioid drugs. Despite that risk, under no circumstances should adequate pain relief ever be withheld simply because an opioid exhibits potential for abuse or because legislative controls complicate the process of prescribing narcotics. Furthermore, certain principles can be observed by the clinician to minimize problems presented by tolerance and dependence when using opioid analgesics: • Establish therapeutic goals before starting opioid therapy. This tends to limit the potential for physical dependence. The patient and his or her family should be included in this process. • Once an effective dose is established, attempt to limit dosage to this level. This goal is facilitated by use of a written treatment contract that specifically prohibits early refills and having multiple prescribing physicians. • Non-opioid analgesics—especially in chronic management—consider using other types of analgesics or compounds exhibiting less pronounced withdrawal symptoms on discontinuance. • Frequently evaluate continuing analgesic therapy and the patient’s need for opioids. B. Diagnosis and Treatment of Opioid Overdosage Intravenous injection of naloxone dramatically reverses coma due to opioid overdose but not that due to other CNS depressants. Use of


the antagonist should not, of course, delay the institution of other therapeutic measures, especially respiratory support. (See also The Opioid Antagonists, below, and Chapter 58.) C. Contraindications and Cautions in Therapy 1. Use of pure agonists with weak partial agonists—When a weak partial agonist such as pentazocine is given to a patient also receiving a full agonist (eg, morphine), there is a risk of diminishing analgesia or even inducing a state of withdrawal; thus combining a full agonist with partial agonist opioids should be avoided. 2. Use in patients with head injuries—Carbon dioxide retention caused by respiratory depression results in cerebral vasodilation. In patients with elevated intracranial pressure, this may lead to lethal alterations in brain function. 3. Use during pregnancy—In pregnant women who are chronically using opioids, the fetus may become physically dependent in utero and manifest withdrawal symptoms in the early postpartum period. A daily dose as small as 6 mg of heroin (or equivalent) taken by the mother can result in a mild withdrawal syndrome in the infant, and twice that much may result in severe signs and symptoms, including irritability, shrill crying, diarrhea, or even seizures. Recognition of the problem is aided by a careful history and physical examination. When withdrawal symptoms are judged to be relatively mild, treatment is aimed at control of these symptoms using such drugs as diazepam; with more severe withdrawal, camphorated tincture of opium (paregoric; 0.4 mg of morphine/mL) in an oral dose of 0.12–0.24 mL/kg is used. Oral doses of methadone (0.1–0.5 mg/kg) have also been used. 4. Use in patients with impaired pulmonary function—In patients with borderline respiratory reserve, the depressant properties of the opioid analgesics may lead to acute respiratory failure. 5. Use in patients with impaired hepatic or renal function—Because morphine and its congeners are metabolized primarily in the liver, their use in patients in prehepatic coma may be questioned. Half-life is prolonged in patients with impaired renal function, and morphine and its active glucuronide metabolite may accumulate; dosage can often be reduced in such patients. 6. Use in patients with endocrine disease—Patients with adrenal insufficiency (Addison’s disease) and those with hypothyroidism (myxedema) may have prolonged and exaggerated responses to opioids.

Drug Interactions Because seriously ill or hospitalized patients may require a large number of drugs, there is always a possibility of drug interactions when the opioid analgesics are administered. Table 31–5 lists some of these drug interactions and the reasons for not combining the named drugs with opioids. TABLE 31–5 Opioid drug interactions.


SPECIFIC AGENTS The following section describes the most important and widely used opioid analgesics, along with features peculiar to specific agents. Data about doses approximately equivalent to 10 mg of intramuscular morphine, oral versus parenteral efficacy, duration of analgesia, and intrinsic activity (maximum efficacy) are presented in Table 31–2.

STRONG AGONISTS Phenanthrenes Morphine, hydromorphone, and oxymorphone are strong agonists useful in treating severe pain. These prototypic agents have been described in detail above.

Heroin (diamorphine, diacetylmorphine) is potent and fast-acting, but its use is prohibited in the USA and Canada. In recent years, there has been considerable agitation to revive its use. However, double-blind studies have not supported the claim that heroin is more effective than morphine in relieving severe chronic pain, at least when given by the intramuscular route.

Phenylheptylamines Methadone has undergone a dramatic revival as a potent and clinically useful analgesic. It can be administered by the oral, intravenous,


subcutaneous, spinal, and rectal routes. It is well absorbed from the gastrointestinal tract and its bioavailability far exceeds that of oral morphine.

Methadone is not only a potent μ-receptor agonist but its racemic mixture of D- and L-methadone isomers can also block both NMDA receptors and monoaminergic reuptake transporters. These nonopioid receptor properties may help explain its ability to relieve difficult-to-treat pain (neuropathic, cancer pain), especially when a previous trial of morphine has failed. In this regard, when analgesic tolerance or intolerable side effects have developed with the use of increasing doses of morphine or hydromorphone, “opioid rotation” to methadone has provided superior analgesia at 10–20% of the morphine-equivalent daily dose. In contrast to its use in suppressing symptoms of opioid withdrawal, use of methadone as an analgesic typically requires administration at intervals of no more than 8 hours. However, given methadone’s highly variable pharmacokinetics and long half-life (25–52 hours), initial administration should be closely monitored to avoid potentially harmful adverse effects, especially respiratory depression. Because methadone is metabolized by CYP3A4 and CYP2B6 isoforms in the liver, inhibition of its metabolic pathway or hepatic dysfunction has also been associated with overdose effects, including respiratory depression or, more rarely, prolonged QT-based cardiac arrhythmias. Methadone is widely used in the treatment of opioid abuse. Tolerance and physical dependence develop more slowly with methadone than with morphine. The withdrawal signs and symptoms occurring after abrupt discontinuance of methadone are milder, although more prolonged, than those of morphine. These properties make methadone a useful drug for detoxification and for maintenance of the chronic relapsing heroin addict. For detoxification of a heroin-dependent addict, low doses of methadone (5–10 mg orally) are given two or three times daily for 2 or 3 days. Upon discontinuing methadone, the addict experiences a mild but endurable withdrawal syndrome. For maintenance therapy of the opioid recidivist, tolerance to 50–100 mg/d of oral methadone may be deliberately produced; in this state, the addict experiences cross-tolerance to heroin, which prevents most of the addiction-reinforcing effects of heroin. One rationale of maintenance programs is that blocking the reinforcement obtained from abuse of illicit opioids removes the drive to obtain them, thereby reducing criminal activity and making the addict more amenable to psychiatric and rehabilitative therapy. The pharmacologic basis for the use of methadone in maintenance programs is sound and the sociologic basis is rational, but some methadone programs fail because nonpharmacologic management is inadequate. The concurrent administration of methadone to heroin addicts known to be recidivists has been questioned because of the increased risk of overdose death secondary to respiratory arrest. As the number of patients prescribed methadone for persistent pain has increased, so, too, has the incidence of accidental overdose and complications related to respiratory depression. Variability in methadone metabolism, protein binding, distribution, and nonlinear opioid dose conversion all play a role in adverse events. Buprenorphine, a partial μreceptor agonist with long-acting properties, has been found to be effective in opioid detoxification and maintenance programs and is presumably associated with a lower risk of such overdose fatalities.

Phenylpiperidines Fentanyl is one of the most widely used agents in the family of synthetic opioids. The fentanyl subgroup now includes sufentanil, alfentanil, and remifentanil in addition to the parent compound, fentanyl.


These opioids differ mainly in their potency and biodisposition. Sufentanil is five to seven times more potent than fentanyl. Alfentanil is considerably less potent than fentanyl, but acts more rapidly and has a markedly shorter duration of action. Remifentanil is metabolized very rapidly by blood and nonspecific tissue esterases, making its pharmacokinetic and pharmacodynamic half-lives extremely short. Such properties are useful when these compounds are used in anesthesia practice. Although fentanyl is now the predominant analgesic in the phenylpiperidine class, meperidine continues to be used. This older opioid has significant antimuscarinic effects, which may be a contraindication if tachycardia would be a problem. Meperidine is also reported to have a negative inotropic action on the heart. In addition, it has the potential for producing seizures secondary to accumulation of its metabolite, normeperidine, in patients receiving high doses or with concurrent renal failure. Given this undesirable profile, use of meperidine as a first-line analgesic is becoming increasingly rare.

Morphinans Levorphanol is a synthetic opioid analgesic closely resembling morphine that has μ-, δ-, and κ-opioid agonist actions, serotoninnorepinephrine reuptake inhibition, and NMDA receptor antagonist properties.

MILD TO MODERATE AGONISTS Phenanthrenes Codeine, dihydrocodeine, and hydrocodone have lower binding affinity to μ-opioid receptors than morphine and often have adverse effects that limit the maximum tolerated dose when one attempts to achieve analgesia comparable to that of morphine. Oxycodone is more potent and is prescribed alone in higher doses as immediate-release or controlled-release forms for the treatment of moderate to severe pain. Combinations of hydrocodone or oxycodone with acetaminophen are the predominant formulations of orally administered analgesics in the United States for the treatment of mild to moderate pain. However, there has been a large increase in the use of controlled-release oxycodone at the highest dose range. An intravenous formulation of oxycodone is available outside the United States. Since each controlled-release tablet of oxycodone contains a large quantity of oxycodone to allow for prolonged action, those intent on abusing the old formulation have extracted crushed tablets and injected high doses, resulting in abuse and possible fatal overdose. In 2010, the FDA approved a new formulation of the controlled-release form of oxycodone that reportedly prevents the tablets from being cut, broken, chewed, crushed, or dissolved to release more oxycodone. It is hoped that this new formulation will lead to less abuse by snorting or injection. The FDA is now requiring a Risk Evaluation and Mitigation Strategy (REMS) that will include the issuance of a medication guide to patients and a requirement for prescriber education regarding the appropriate use of opioid analgesics in the treatment of pain. (See Box: Educating Opioid Prescribers.)

Phenylheptylamines


Propoxyphene is chemically related to methadone but has extremely low analgesic activity. Its low efficacy makes it unsuitable, even in combination with aspirin, for severe pain. The increasing incidence of deaths associated with its use and misuse caused it to be withdrawn in the United States.

Phenylpiperidines Diphenoxylate and its metabolite, difenoxin, are not used for analgesia but for the treatment of diarrhea. They are scheduled for minimal control (difenoxin is Schedule IV, diphenoxylate Schedule V; see inside front cover) because the likelihood of their abuse is remote. The poor solubility of the compounds limits their use for parenteral injection. As antidiarrheal drugs, they are used in combination with atropine. The atropine is added in a concentration too low to have a significant antidiarrheal effect but is presumed to further reduce the likelihood of abuse. Loperamide is a phenylpiperidine derivative used to control diarrhea. Due to action on peripheral μ-opioid receptors and lack of effect on CNS receptors, investigations are ongoing as to whether it could be an effective analgesic. Its potential for abuse is considered very low because of its limited access to the brain. It is therefore available without a prescription.

Educating Opioid Prescribers The treatment of pain is a difficult clinical-pharmacologic problem, and prescribers of opioids have often failed to appreciate this difficulty. As a result, there have been large increases of drug abuse cases in the USA and a nearly fourfold increase in overdose deaths due to prescription opioids between 1999 and the present. These statistics have prompted the Food and Drug Administration to formulate plans for opioid manufacturers to provide training for all opioid prescribers. The FDA is working to devise methods by which this training would be mandatory for all prescribers and would emphasize the thorough understanding of opioid clinical pharmacology with special education about long-acting and extended-release formulations. The educational emphasis on the longacting and sustained-release formulations (eg, methadone, oxycodone) reflects their association with skyrocketing morbidity and mortality. The usual dose with all of these antidiarrheal agents is two tablets to start and then one tablet after each diarrheal stool.

OPIOIDS WITH MIXED RECEPTOR ACTIONS Care should be taken not to administer any partial agonist or drug with mixed opioid receptor actions to patients receiving pure agonist drugs because of the unpredictability of both drugs’ effects; reduction of analgesia or precipitation of an explosive abstinence syndrome may result.

Phenanthrenes As noted above, buprenorphine is a potent and long-acting phenanthrene derivative that is a partial μ-receptor agonist (low intrinsic activity) and an antagonist at the δ and κ receptors and is therefore referred to as a mixed agonist-antagonist. Although buprenorphine is used as an analgesic, it can antagonize the action of more potent μ agonists such as morphine. Buprenorphine also binds to ORL1, the orphanin receptor. Whether this property also participates in opposing μ receptor function is under study. Administration by the sublingual route is preferred to avoid significant first-pass effect. Buprenorphine’s long duration of action is due to its slow dissociation from μ receptors. This property renders its effects resistant to naloxone reversal. Buprenorphine was approved by the FDA in 2002 for the management of opioid dependence and studies suggest it is as effective as methadone for the management of opioid withdrawal and detoxification in programs that include counseling, psychosocial support, and direction by physicians qualified under the Drug Addiction Treatment Act. In contrast to methadone, high-dose administration of buprenorphine results in a μ-opioid antagonist action, limiting its properties of analgesia and respiratory depression. However, buprenorphine formulations can still cause serious respiratory depression and death, particularly when extracted and injected intravenously in combination with benzodiazepines or used with other CNS depressants (ie, sedatives, antipsychotics, or alcohol). Buprenorphine is also available combined with naloxone, a pure μ-opioid antagonist (as Suboxone), to help prevent its diversion for illicit intravenous abuse. A slow-release transdermal patch preparation that releases drug over a 1-week period is also available (Butrans). Psychotomimetic effects, with hallucinations, nightmares, and anxiety, have been reported after use of drugs with mixed agonist-antagonist actions. Pentazocine (a benzomorphan) and nalbuphine are other examples of opioid analgesics with mixed agonist-antagonist properties. Nalbuphine is a strong κ-receptor agonist and a partial μ-receptor antagonist; it is given parenterally. At higher doses there seems to be a definite ceiling—not noted with morphine—to the respiratory depressant effect. Unfortunately, when respiratory depression does occur, it may be relatively resistant to naloxone reversal due to its greater affinity for the receptor than naloxone.


Morphinans Butorphanol produces analgesia equivalent to nalbuphine but appears to produce more sedation at equianalgesic doses. Butorphanol is considered to be predominantly a κ agonist. However, it may also act as a partial agonist or antagonist at the μ receptor.

Benzomorphans Pentazocine is a κ agonist with weak μ-antagonist or partial agonist properties. It is the oldest mixed agent available. It may be used orally or parenterally. However, because of its irritant properties, the injection of pentazocine subcutaneously is not recommended.

MISCELLANEOUS Tramadol is a centrally acting analgesic whose mechanism of action is predominantly based on blockade of serotonin reuptake. Tramadol has also been found to inhibit norepinephrine transporter function. Because its analgesic effect is only partially antagonized by naloxone, it is thought to not depend on its low-affinity binding to the μ receptor for therapeutic activity. The recommended dosage is 50– 100 mg orally four times daily. Toxicity includes association with seizures; the drug is relatively contraindicated in patients with a history of epilepsy and for use with other drugs that lower the seizure threshold. Another serious risk is the development of serotonin syndrome, especially if selective serotonin reuptake inhibitor antidepressants are being administered (see Chapter 16). Other adverse effects include nausea and dizziness, but these symptoms typically abate after several days of therapy. No clinically significant effects on respiration or the cardiovascular system have thus far been reported. Given the fact that the analgesic action of tramadol is largely independent of μreceptor action, tramadol may serve as an adjunct with pure opioid agonists in the treatment of chronic neuropathic pain. Tapentadol is an analgesic with modest μ-opioid receptor affinity and significant norepinephrine reuptake-inhibiting action. In animal models, its analgesic effects were only moderately reduced by naloxone but strongly reduced by an α2 -adrenoceptor antagonist. Furthermore, its binding to the norepinephrine transporter (NET, see Chapter 6) was stronger than that of tramadol, whereas its binding to the serotonin transporter (SERT) was less than that of tramadol. Tapentadol was approved in 2008 and has been shown to be as effective as oxycodone in the treatment of moderate to severe pain but with a reduced profile of gastrointestinal complaints such as nausea. Tapentadol carries risk for seizures in patients with seizure disorders and for the development of serotonin syndrome. It is unknown how tapentadol compares in clinical utility to tramadol or other analgesics whose mechanism of action is not based primarily on opioid receptor pharmacology.

ANTITUSSIVES The opioid analgesics are among the most effective drugs available for the suppression of cough. This effect is often achieved at doses below those necessary to produce analgesia. The receptors involved in the antitussive effect appear to differ from those associated with the other actions of opioids. For example, the antitussive effect is also produced by stereoisomers of opioid molecules that are devoid of analgesic effects and addiction liability (see below). The physiologic mechanism of cough is complex, and little is known about the specific mechanism of action of the opioid antitussive drugs. It appears likely that both central and peripheral effects play a role. The opioid derivatives most commonly used as antitussives are dextromethorphan, codeine, levopropoxyphene, and noscapine (levopropoxyphene and noscapine are not available in the USA). They should be used with caution in patients taking monoamine oxidase inhibitors (Table 31–5). Antitussive preparations usually also contain expectorants to thin and liquefy respiratory secretions. Importantly, due to increasing reports of death in young children taking dextromethorphan in formulations of over-the-counter “cold/cough” medications, its use in children younger than 6 years of age has been banned by the FDA. Moreover, because of variations in the metabolism of codeine, its use for any purpose in young children is being reconsidered. Dextromethorphan is the dextrorotatory stereoisomer of a methylated derivative of levorphanol. It is purported to be free of addictive properties and produces less constipation than codeine. The usual antitussive dose is 15–30 mg three or four times daily. It is available in many over-the-counter products. Dextromethorphan has also been found to enhance the analgesic action of morphine and presumably other μ-receptor agonists. However, abuse of its purified (powdered) form has been reported to lead to serious adverse events including death. Codeine, as noted, has a useful antitussive action at doses lower than those required for analgesia. Thus, 15 mg is usually sufficient to relieve cough. Levopropoxyphene is the stereoisomer of the weak opioid agonist dextropropoxyphene. It is devoid of opioid effects, although sedation has been described as a side effect. The usual antitussive dose is 50–100 mg every 4 hours.

THE OPIOID ANTAGONISTS


The pure opioid antagonist drugs naloxone, naltrexone, and nalmefene are morphine derivatives with bulkier substituents at the N17 position. These agents have a relatively high affinity for μ-opioid binding sites. They have lower affinity for the other receptors but can also reverse agonists at δ and κ sites.

Pharmacokinetics Naloxone is usually given by injection and has a short duration of action (1–2 hours) when given by this route. Metabolic disposition is chiefly by glucuronide conjugation like that of the agonist opioids with free hydroxyl groups. Naltrexone is well absorbed after oral administration but may undergo rapid first-pass metabolism. It has a half-life of 10 hours, and a single oral dose of 100 mg blocks the effects of injected heroin for up to 48 hours. Nalmefene, the newest of these agents, is a derivative of naltrexone but is available only for intravenous administration. Like naloxone, nalmefene is used for opioid overdose but has a longer half-life (8–10 hours).

Pharmacodynamics When given in the absence of an agonist drug, these antagonists are almost inert at doses that produce marked antagonism of agonist opioid effects. When given intravenously to a morphine-treated subject, the antagonist completely and dramatically reverses the opioid effects within 1–3 minutes. In individuals who are acutely depressed by an overdose of an opioid, the antagonist effectively normalizes respiration, level of consciousness, pupil size, bowel activity, and awareness of pain. In dependent subjects who appear normal while taking opioids, naloxone or naltrexone almost instantaneously precipitates an abstinence syndrome. There is no tolerance to the antagonistic action of these agents, nor does withdrawal after chronic administration precipitate an abstinence syndrome.

Clinical Use Naloxone is a pure antagonist and is preferred over older weak agonist-antagonist agents that had been used primarily as antagonists, eg, nalorphine and levallorphan. The major application of naloxone is in the treatment of acute opioid overdose (see also Chapter 58). It is very important that the relatively short duration of action of naloxone be borne in mind, because a severely depressed patient may recover after a single dose of naloxone and appear normal, only to relapse into coma after 1–2 hours. The usual initial dose of naloxone is 0.1–0.4 mg intravenously for life-threatening respiratory and CNS depression. Maintenance is with the same drug, 0.4–0.8 mg given intravenously, and repeated whenever necessary. In using naloxone in the severely opioiddepressed newborn, it is important to start with doses of 5–10 mcg/kg and to consider a second dose of up to a total of 25 mcg/kg if no response is noted. Low-dose naloxone (0.04 mg) has an increasing role in the treatment of adverse effects that are commonly associated with intravenous or epidural opioids. Careful titration of the naloxone dosage can often eliminate the itching, nausea, and vomiting while sparing the analgesia. For this purpose, oral naloxone, and modified analogs of naloxone and naltrexone, have been approved by the FDA. These include methylnaltrexone bromide for the treatment of constipation in patients with late-stage advanced illness and alvimopan for the treatment of postoperative ileus following bowel resection surgery. Methylnaltrexone has a quaternary amine preventing it from crossing the blood-brain barrier. Alvimopan has a high affinity for peripheral μ receptors and does not impair the central effects of μ-opioid agonists. The principal mechanism for the selective therapeutic effect of these agents is peripheral enteric μreceptor antagonism with minimal CNS penetration. Because of its long duration of action, naltrexone has been proposed as a maintenance drug for addicts in treatment programs. A single dose given on alternate days blocks virtually all of the effects of a dose of heroin. It might be predicted that this approach to rehabilitation would not be popular with a large percentage of drug users unless they are motivated to become drug-free. A related use is in combination with morphine sulfate in a controlled-release formulation (Embeda) in which 20–100 mg of morphine is slowly released over 8–12 hours or longer for the control of prolonged postoperative pain. Naltrexone, 0.4–4 mg, is sequestered in the center of the


formulation pellets and is present to prevent the abuse of the morphine (by grinding and extraction of the morphine from the capsules). There is evidence that naltrexone decreases the craving for alcohol in chronic alcoholics by increasing baseline β-endorphin release, and it has been approved by the FDA for this purpose (see Chapter 23). Naltrexone also facilitates abstinence from nicotine (cigarette smoking) with reduced weight gain. In fact, a combination of naltrexone plus bupropion (Chapter 16) may also offer an effective and synergistic strategy for weight loss. If current trials demonstrate cardiovascular safety during prolonged use, this and other weight-loss medications combined with naltrexone may eventually win FDA approval.

SUMMARY Opioids, Opioid Substitutes, and Opioid Antagonists



PREPARATIONS AVAILABLE*



REFERENCES Angst MS, Clark JD: Opioid-induced hyperalgesia. Anesthesiology 2006;104:570. Anton RF: Naltrexone for the management of alcohol dependence. N Eng J Med 2008;359:715. Basbaum AI et al: Cellular and molecular mechanisms of pain. Cell 2009;139:267. Basbaum AI, Jessel T : T he perception of pain. In: Kandel ER et al (editors): Principles of Neural Science, 4th ed. McGraw-Hill, 2000. Benedetti C, Premuda L: T he history of opium and its derivatives. In: Benedetti C et al (editors): Advances in Pain Research and Therapy, vol 14. Raven Press, 1990. Bolan EA, T allarida RJ, Pasternak GW: Synergy between mu opioid ligands: Evidence for functional interactions among mu opioid receptor subtypes. J Pharmacol Exp T her 2002;303:557. Chu LF, Angst MS, Clark D: Opioid-induced hyperalgesia in humans: Molecular mechanisms and clinical considerations. Clin J Pain 2008;24:479. Curran MP et al: Alvimopan. Drugs 2008;68:2011. Dahan A et al: Sex-specific responses to opiates: Animal and human studies. Anesth Analg 2008;107:83. Davis MP, Walsh D: Methadone for relief of cancer pain: A review of pharmacokinetics, pharmacodynamics, drug interactions and protocols of administration. Support Care Cancer 2001;9:73. Ferner RE, Daniels AM: Office-based treatment of opioid-dependent patients. N Engl J Med 2003;348:81. Ferrante FM: Principles of opioid pharmacotherapy: Practical implications of basic mechanisms. J Pain Symptom Manage 1996;11:265. Fields HL, Basbaum AI: Central nervous system mechanisms of pain modulation. In: Wall PD, Melzack R (editors): Textbook of Pain. Churchill Livingstone, 1999. Fillingim RB, Gear RW: Sex differences in opioid analgesia: Clinical and experimental findings. Eur J Pain 2004;8:413. Fischer BD, Carrigan KA, Dykstra LA: Effects of N-methyl-D-aspartate receptor antagonists on acute morphine-induced and L-methadone-induced antinociception in mice. J Pain 2005;6:425. Goldman D, Barr CS: Restoring the addicted brain. N Engl J Med 2002;347:843. Inui S: Nalfurafine hydrochloride for the treatment of pruritus. Expert Opin Pharmacother 2012;13:1507. Joly V et al: Remifentanil-induced postoperative hyperalgesia and its prevention with small-dose ketamine. Anesthesiology 2005;103:147. Julius D, Basbaum AI: Molecular mechanisms of nociception. Nature 2001;413:203. Kalso E et al: No pain, no gain: Clinical excellence and scientific rigour—lessons learned from IA morphine. Pain 2002;98:269. Kiefer BL: Opioids: First lessons from knockout mice. T rends Pharmacol Sci 1999;20:19. Kim JA et al: Morphine-induced receptor endocytosis in a novel knockin mouse reduces tolerance and dependence. Curr Biol 2008;18:129. King T et al: Role of NK-1 neurotransmission in opioid-induced hyperalgesia. Pain 2005;116:276. Lai J et al: Pronociceptive actions of dynorphin via bradykinin receptors. Neurosci Lett 2008;437:175. Lambert DG: T he nociceptin/orphanin FQ receptor: A target with broad therapeutic potential. Nat Rev Drug Discov 2008;7:694. Laughlin T M, Larson AA, Wilcox GL: Mechanisms of induction of persistent nociception by dynorphin. J Pharmacol Exp T her 2001;299:6. Liaw WI et al: Distinct expression of synaptic NR2A and NR2B in the central nervous system and impaired morphine tolerance and physical dependence in mice deficient in postsynaptic density-93 protein. Mol Pain 2008;4:45. Liu XY et al: Unidirectional cross-activation of GRPR by MOR1D uncouples itch and analgesia induced by opioids. Cell 2011;147:447. McGaraughty S, Heinricher MM: Microinjection of morphine into various amygdaloid nuclei differentially affects nociceptive responsiveness and RVM neuronal activity. Pain 2002;96:153. Mercadante S, Arcuri E: Opioids and renal function. J Pain 2004;5:2. Meunier J, Mouledous L, T opham CM: T he nociceptin (ORL1) receptor: Molecular cloning and functional architecture. Peptides 2000;21:893. Mitchell JM, Basbaum AI, Fields HL: A locus and mechanism of action for associative morphine tolerance. Nat Neurosci 2000;3:47. Okie S: A flood of opioids, a rising tide of deaths. N Engl J Med 2010;363:1981. Pan YX: Diversity and complexity of the mu opioid receptor gene: Alternate pre-mRNA splicing and promoters. DNA Cell Biol 2005;24:736. Reimann F et al: Pain perception is altered by a nucleotide polymorphism in SCN9A. Proc Natl Acad Sci USA 2010;107:5148. Reynolds SM et al: T he pharmacology of cough. T rends Pharmacol Sci 2004;25:569. Rittner HL, Brack A, Stein C: Pain and the immune system. Br J Anaesth 2008;101:40. Scherrer G et al: Dissociation of the opioid receptor mechanisms that control mechanical and heat pain. Cell 2009;137:1148. Skarke C, Geisslinger G, Lotsch J: Is morphine-3-glucuronide of therapeutic relevance? Pain 2005;116:177. Smith MT : Differences between and combinations of opioids revisited. Curr Opin Anaesthesiol 2008;21:596. Smith MT : Neuroexcitatory effects of morphine and hydromorphone: Evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol 2000;27:524. Stein C, Schafer M, Machelska H: Attacking pain at its source: New perspectives on opioids. Nat Med 2003;9:1003. Vanderah T W et al: Mechanisms of opioid-induced pain and antinociceptive tolerance: Descending facilitation and spinal dynorphin. Pain 2001;92:5. Waldhoer M et al: A heterodimer-selective agonist shows in vivo relevance of G protein-coupled receptor dimers. Proc Natl Acad Sci USA 2005;102:9050. Wang Z et al: Pronociceptive actions of dynorphin maintain chronic neuropathic pain. J Neurosci 2001;21:1779. Wild JE et al: Long-term safety and tolerability of tapentadol extended release for the management of chronic low back pain or osteoarthritis pain. Pain Pract 2010;10:416. Williams JT , Christie MJ, Manzoni O: Cellular and synaptic adaptations mediating opioid dependence. Physiol Rev 2001;81:299. Woolf CJ, Salter MW: Neuronal plasticity: Increasing the gain in pain. Science 2000;288:1765. Zhao GM et al: Profound spinal tolerance after repeated exposure to a highly selective mu-opioid peptide agonist: Role of delta-opioid receptors. J Pharmacol Exp T her 2002;302:188. Zubieta JK et al: Regional mu opioid receptor regulation of sensory and affective dimensions of pain. Science 2001;293:311.

CASE STUDY ANSWER


In this case, the treatment of severe pain should be managed with the administration of a potent intravenous opioid analgesic such as morphine, hydromorphone, or fentanyl. Before an additional dose of an opioid analgesic is administered, it is expected that the patient will require frequent reevaluation of both the severity of his pain and the presence of potential side effects. Given his history of pulmonary disease, he is also at increased risk of developing respiratory depression. Frequent reevaluation of his level of consciousness, respiratory rate, fractional oxygen saturation, and other vital parameters can help achieve the goal of pain relief and minimize respiratory depression. Concurrent use of sedative agents such as benzodiazepines should be avoided if possible and proceed only with great caution.


_______________ * In memory of Walter (Skip) Way, MD.


CHAPTER

32 Drugs of Abuse Christian Lüscher, MD

CASE STUDY Mr V, a 47-year-old man, was recently promoted as a director of a transportation company. A routine inspection of the books shows that a large sum of money is missing. Subsequent investigation finds that Mr V has been spending more than $20,000 a month to buy cocaine; currently he consumes 2–3 g/d. He also drinks several beers each day and 5–8 shots of vodka in the evening. He spends weekend nights in clubs, where he often consumes 2–3 pills of ecstasy. He began using drugs at age 18; during parties he mostly smoked cannabis (5–6 joints per weekend), but also tried cocaine. This “recreational use” came to an abrupt halt when he married at age 27 and entered a professional training program that allowed him to obtain his current job, now jeopardized by his cocaine use. Is Mr V addicted, dependent, or both? What is the reason for the use of several different addictive drugs at the same time?

Drugs are abused (used in ways that are not medically approved) because they cause strong feelings of euphoria or alter perception. However, repetitive exposure induces widespread adaptive changes in the brain. As a consequence, drug use may become compulsive— the hallmark of addiction.

BASIC NEUROBIOLOGY OF DRUG ABUSE DEPENDENCE VERSUS ADDICTION Recent neurobiologic research has led to the conceptual and mechanistic separation of “dependence” and “addiction.” The older term “physical dependence” is now denoted as dependence, whereas “psychological dependence” is more simply called addiction. Every addictive drug causes its own characteristic spectrum of acute effects, but all have in common that they induce strong feelings of euphoria and reward. With repetitive exposure, addictive drugs induce adaptive changes such as tolerance (ie, escalation of dose to maintain effect). Once the abused drug is no longer available, signs of withdrawal become apparent. A combination of such signs, referred to as the withdrawal syndrome, defines dependence. Dependence is not always a correlate of drug abuse—it can also occur with many classes of nonpsychoactive drugs, eg, sympathomimetic vasoconstrictors and bronchodilators, and organic nitrate vasodilators. Addiction, on the other hand, consists of compulsive, relapsing drug use despite negative consequences, at times triggered by cravings that occur in response to contextual cues (see Box: Animal Models in Addiction Research). Although dependence invariably occurs with chronic exposure, only a small percentage of subjects develop a habit, lose control, and become addicted. For example, very few patients who receive opioids as analgesics desire the drug after withdrawal. And only one person out of six becomes addicted within 10 years of first use of cocaine. Conversely, relapse is very common in addicts after a successful withdrawal when, by definition, they are no longer dependent.

ADDICTIVE DRUGS INCREASE THE LEVEL OF DOPAMINE: REINFORCEMENT To understand the long-term changes induced by drugs of abuse, their initial molecular and cellular targets must be identified. A combination of approaches in animals and humans, including functional imaging, has revealed the mesolimbic dopamine system as the prime target of addictive drugs. This system originates in the ventral tegmental area (VTA), a tiny structure at the tip of the brainstem, which projects to the nucleus accumbens, the amygdala, the hippocampus, and the prefrontal cortex (Figure 32–1). Most projection neurons of the VTA are dopamine-producing neurons. When the dopamine neurons of the VTA begin to fire in bursts, large quantities of


dopamine are released in the nucleus accumbens and the prefrontal cortex. Early animal studies pairing electrical stimulation of the VTA with operant responses (eg, lever pressing) that result in strong reinforcement established the central role of the mesolimbic dopamine system in reward processing. Direct application of drugs into the VTA also acts as a strong reinforcer, and systemic administration of drugs of abuse causes release of dopamine. Even selective activation of dopamine neurons is sufficient to elicit behavioral changes typically observed with addictive drugs. These very selective interventions use optogenetic methods. Blue light is delivered in a freely moving mouse through light guides to activate channelrhodopsin, a light-gated cation channel that is artificially expressed in dopamine neurons. As a result, mice will self-administer blue light; pairing light activation of VTA dopamine neurons with a specific environment establishes a long-lasting place preference. Conversely using inhibitory optogenetic effectors or activation of inhibitory neurons upstream causes aversion.

FIGURE 32–1 Major connections of the mesolimbic dopamine system in the brain. Schematic diagram of brain sections illustrating that the dopamine projections originate in the ventral tegmental area and target the nucleus accumbens, prefrontal cortex, amygdala, and


hippocampus. The dashed lines on the sagittal section indicate where the horizontal and coronal sections were made. As a general rule, all addictive drugs activate the mesolimbic dopamine system. The behavioral significance of this increase of dopamine is still debated. An appealing hypothesis is that mesolimbic dopamine codes for the difference between expected and actual reward and thus constitutes a strong learning signal (see Box: The Dopamine Hypothesis of Addiction). Since each addictive drug has a specific molecular target that engages distinct cellular mechanisms to activate the mesolimbic system, three classes can be distinguished: A first group binds to G io protein-coupled receptors, a second group interacts with ionotropic receptors or ion channels, and a third group targets the dopamine transporter (Table 32–1 and Figure 32–2). G proteincoupled receptors (GPCRs) of the Gio family inhibit neurons through postsynaptic hyperpolarization and presynaptic regulation of transmitter release. In the VTA, the action of these drugs is preferentially on the γ-aminobutyric acid (GABA) neurons that act as local inhibitory interneurons. Addictive drugs that bind to ionotropic receptors and ion channels can have combined effects on dopamine neurons and GABA neurons, eventually leading to enhanced release of dopamine. Finally, addictive drugs that interfere with monoamine transporters block reuptake or stimulate nonvesicular release of dopamine, causing an accumulation of extracellular dopamine in target structures. Since neurons of the VTA also express somatodendritic transporters, which normally clear dopamine released by the dendrites, class 3 drugs also increase dopamine level in the VTA. Although drugs of this class also affect transporters of other monoamines (norepinephrine, serotonin), action on the dopamine transporter remains central for addiction. This is consistent with the observations that antidepressants that block serotonin and norepinephrine uptake, but not dopamine uptake, do not cause addiction even after prolonged use. TABLE 32–1 The mechanistic classification of drugs of abuse.1



FIGURE 32–2 Neuropharmacologic classification of addictive drugs by primary target (see text and Table 32–1). DA, dopamine; GABA, γ-aminobutyric acid; GHB, γ-hydroxybutyric acid; GPCRs, G protein-coupled receptors; THC, Δ9 -tetrahydrocannabinol.

Animal Models in Addiction Research Many of the recent advances in addiction research have been made possible by the use of animal models. Since drugs of abuse are not only rewarding but also reinforcing, an animal will learn a behavior (eg, press a lever) when paired with drug administration. In such a self-administration paradigm, the number of times an animal is willing to press the lever in order to obtain a single dose reflects the strength of reinforcement and is therefore a measure of the rewarding properties of a drug. Observing withdrawal signs specific for rodents (eg, escape jumps or “wet-dog” shakes after abrupt termination of chronic morphine administration) allows the quantification of dependence. Behavioral tests for addiction in the rodent have proven difficult to develop, and so far no test fully captures the complexity of the disease. However, it is possible to model core components of addiction; for example, by monitoring behavioral sensitization and conditioned place preference. In the first test, an increase in locomotor activity is observed with intermittent drug exposure. The latter tests for the preference of a particular environment associated with drug exposure by measuring the time an animal spends in the compartment where a drug was received compared with the compartment where only saline was injected (conditioned place preference). Both tests have in common that they are sensitive to cue-conditioned effects of addictive drugs. Subsequent exposures to the environment without the drug lead to extinction of the place preference, which can be reinstated with a low dose of the drug or the presentation of a conditioned stimulus. These persistent changes serve as a model of relapse and have been linked to synaptic plasticity of excitatory transmission in the ventral tegmental area, nucleus accumbens, and prefrontal cortex (see also Box: The Dopamine Hypothesis of Addiction). More sophisticated tests rely on self-administration of the drug, in which a rat or a mouse has to press a lever in order to obtain an injection of, for example, cocaine. Once the animal has learned the association with a conditioned stimulus (eg, light or brief sound), the simple presentation of the cue elicits drug seeking. Prolonged self-administration of addictive drugs over months leads to behaviors in rats that closely resemble human addiction. Such “addicted” rodents are very strongly motivated to seek cocaine, continue looking for the drug even when no longer available, and self-administer cocaine in spite of negative consequences, such as an electric foot shock. These findings suggest that addiction is a disease that does not respect species boundaries.

DEPENDENCE: TOLERANCE & WITHDRAWAL


With chronic exposure to addictive drugs, the brain shows signs of adaptation. For example, if morphine is used at short intervals, the dose has to be progressively increased over the course of several days to maintain rewarding or analgesic effects. This phenomenon is called tolerance. It may become a serious problem because of increasing side effects—eg, respiratory depression—that do not show as much tolerance and may lead to fatalities associated with overdose. Tolerance to opioids may be due to a reduction of the concentration of a drug or a shorter duration of action in a target system (pharmacokinetic tolerance). Alternatively, it may involve changes of μ-opioid receptor function (pharmacodynamic tolerance). In fact, many μ-opioid receptor agonists promote strong receptor phosphorylation that triggers the recruitment of the adaptor protein β-arrestin, causing G proteins to uncouple from the receptor and to internalize within minutes (see Chapter 2). Since this decreases signaling, it is tempting to explain tolerance by such a mechanism. However, morphine, which strongly induces tolerance, does not recruit β-arrestins and fails to promote receptor internalization. Conversely, other agonists that drive receptor internalization very efficiently induce only modest tolerance. Based on these observations, it has been hypothesized that desensitization and receptor internalization actually protect the cell from overstimulation. In this model, morphine, by failing to trigger receptor endocytosis, disproportionally stimulates adaptive processes, which eventually cause tolerance. Although the molecular identity of these processes is still under investigation, they may be similar to the ones involved in withdrawal (see below). Adaptive changes become fully apparent once drug exposure is terminated. This state is called withdrawal and is observed to varying degrees after chronic exposure to most drugs of abuse. Withdrawal from opioids in humans is particularly strong (described below). Studies in rodents have added significantly to our understanding of the neural and molecular mechanisms that underlie dependence. For example, signs of dependence, as well as analgesia and reward, are abolished in knockout mice lacking the μ-opioid receptor, but not in mice lacking other opioid receptors (δ, κ). Although activation of the μ-opioid receptor initially strongly inhibits adenylyl cyclase, this inhibition becomes weaker after several days of repeated exposure. The reduction of the inhibition of adenylyl cyclase is due to a counter-adaptation of the enzyme system during exposure to the drug, which results in overproduction of cAMP during subsequent withdrawal. Several mechanisms exist for this adenylyl cyclase compensatory response, including up-regulation of transcription of the enzyme. Increased cAMP concentrations in turn strongly activate the transcription factor cyclic AMP response element binding protein (CREB), leading to the regulation of downstream genes. Of the few such genes identified to date, one of the most interesting is the gene for the endogenous κ-opioid ligand dynorphin. The main targets of dynorphin are the presynaptic κ-opioid receptors that regulate the release of dopamine in the nucleus accumbens.

ADDICTION: A DISEASE OF MALADAPTIVE MALADAPTIVE Addiction is characterized by a high motivation to obtain and use a drug despite negative consequences. With time, drug use becomes compulsive (“wanting without liking”). Addiction is a recalcitrant, chronic, and stubbornly relapsing disease that is very difficult to treat. The central problem is that even after successful withdrawal and prolonged drug-free periods, addicted individuals have a high risk of relapsing. Relapse is typically triggered by one of the following three conditions: re-exposure to the addictive drug, stress, or a context that recalls prior drug use. It appears that when paired with drug use, a neutral stimulus may undergo a switch and motivate (“trigger”) addiction-related behavior. This phenomenon may involve synaptic plasticity in the target nuclei of the mesolimbic projection (eg, projections from the medial prefrontal cortex to the neurons of the nucleus accumbens that express the D1 receptors). Several recent studies suggest that the recruitment of the dorsal striatum is responsible for the compulsion. This switch may depend on synaptic plasticity in the nucleus accumbens of the ventral striatum, where mesolimbic dopamine afferents converge with glutamatergic afferents to modulate their function. If dopamine release codes for the prediction error of reward (see Box: The Dopamine Hypothesis of Addiction), pharmacologic stimulation of the mesolimbic dopamine systems will generate an unusually strong learning signal. Unlike natural rewards, addictive drugs continue to increase dopamine even when reward is expected. Such overriding of the prediction error signal may eventually be responsible for the usurping of memory processes by addictive drugs.

The Dopamine Hypothesis of Addiction In the earliest version of the hypothesis described in this chapter, mesolimbic dopamine was believed to be the neurochemical correlate of pleasure and reward. However, during the past decade, experimental evidence has led to several revisions. Phasic dopamine release may actually code for the prediction error of reward rather than the reward itself. This distinction is based on pioneering observations in monkeys that dopamine neurons in the ventral tegmental area (VTA) are most efficiently activated by a reward (eg, a few drops of fruit juice) that is not anticipated. When the animal learns to predict the occurrence of a reward (eg, by pairing it with a stimulus such as a sound), dopamine neurons stop responding to the reward itself (juice), but increase their firing rate when the conditioned stimulus (sound) occurs. Finally, if reward is predicted but not delivered (sound but no juice), dopamine neurons are inhibited below their baseline activity and become silent. In other words, the mesolimbic system continuously scans the reward situation. It increases its activity when reward is larger than expected, and shuts down in the opposite case, thus coding for the prediction error of reward.


Under physiologic conditions the mesolimbic dopamine signal could represent a learning signal responsible for reinforcing constructive behavioral adaptation (eg, learning to press a lever for food). Addictive drugs, by directly increasing dopamine, would generate a strong but inappropriate learning signal, thus hijacking the reward system and leading to pathologic reinforcement. As a consequence, behavior becomes compulsive; that is decisions are no longer planned and under control, but automatic, which is the hallmark of addiction. This appealing hypothesis has been challenged based on the observation that some reward and drug-related learning is still possible in the absence of dopamine. Another intriguing observation is that mice genetically modified to lack the primary molecular target of cocaine, the dopamine transporter DAT, still self-administer the drug. Only when transporters of other biogenic amines are also knocked out does cocaine completely lose its rewarding properties. However, in DAT -/- mice, in which basal synaptic dopamine levels are high, cocaine still leads to increased dopamine release, presumably because other cocaine-sensitive monoamine transporters (NET, SERT) are able to clear some dopamine. When cocaine is given, these transporters are also inhibited and dopamine is again increased. As a consequence of this substitution among monoamine transporters, fluoxetine (a selective serotonin reuptake inhibitor, see Chapter 30) becomes addictive in DAT -/- mice. This concept is supported by newer evidence showing that deletion of the cocaine-binding site on DAT leaves basal dopamine levels unchanged but abolishes the rewarding effect of cocaine. The dopamine hypothesis of addiction has also been challenged by the observation that salient stimuli that are not rewarding (they may actually even be aversive and therefore negative reinforcers) also activate a subpopulation of dopamine neurons in the VTA. The neurons that are activated by aversive stimuli preferentially project to the prefrontal cortex, while the dopamine neurons inhibited by aversive stimuli are those that mostly target the nucleus accumbens. These recent findings suggest that in parallel to the reward system, a system for aversion-learning originates in the VTA. Regardless of the many roles of dopamine under physiologic conditions, all addictive drugs significantly increase its concentration in target structures of the mesolimbic projection. This suggests that high levels of dopamine may actually be at the origin of the adaptive changes that underlie dependence and addiction, a concept that is now supported by novel techniques that allow controlling the activity of dopamine neurons in vivo. In fact manipulations that drive sustained activity of VTA dopamine neurons cause the same cellular adaptations and behavioral changes typically observed with addictive drug exposure. The involvement of learning and memory systems in addiction is also suggested by clinical studies. For example, the role of context in relapse is supported by the report that soldiers who became addicted to heroin during the Vietnam War had significantly better outcomes when treated after their return home, compared with addicts who remained in the environment where they had taken the drug. In other words, cravings may recur at the presentation of contextual cues (eg, people, places, or drug paraphernalia). Current research therefore focuses on the effects of drugs on associative forms of synaptic plasticity, such as long-term potentiation (LTP), which underlie learning and memory (see Box: Synaptic Plasticity & Addiction).

Synaptic Plasticity & Addiction Long-term potentiation (LTP) is a form of experience-dependent synaptic plasticity that is induced by activating glutamate receptors of the N-methyl-D-aspartate (NMDA) type. Since NMDA receptors are blocked by magnesium at negative potentials, their activation requires the concomitant release of glutamate (presynaptic activity) onto a receiving neuron that is depolarized (postsynaptic activity). Correlated pre- and postsynaptic activity durably enhances synaptic efficacy and triggers the formation of new connections. Because associativity is a critical component, LTP has become a leading candidate mechanism underlying learning and memory. LTP can be elicited at glutamatergic synapses of the mesolimbic reward system and is modulated by dopamine. Drugs of abuse could therefore interfere with LTP at sites of convergence of dopamine and glutamate projections (eg, ventral tegmental area [VTA], nucleus accumbens, or prefrontal cortex). Interestingly, exposure to an addictive drug triggers a specific form of synaptic plasticity at excitatory afferents (drug-evoked synaptic plasticity) and potentiates GABAA receptormediated inhibition of the VTA GABA neurons. As a consequence, the excitability of dopamine neurons is increased, the synaptic calcium sources altered, and the rules for subsequent LTP inverted. In the nucleus accumbens, drug-evoked synaptic plasticity appears with some delay and mostly involves the D1 receptor-expressing neurons, which are the ones projecting back to the VTA to control the activity of the GABA neurons. Manipulations in mice that prevent or reverse drug-evoked plasticity in vivo also have effects on persistent changes of drug-associated behavioral sensitization or cue-induced drug seeking, providing more direct evidence for a causal role of synaptic plasticity in drug-adaptive behavior. Non-substance-dependent disorders, such as pathologic gambling and compulsive shopping, share many clinical features of addiction. Several lines of arguments suggest that they also share the underlying neurobiologic mechanisms. This conclusion is supported by the clinical observation that, as an adverse effect of dopamine agonist medication, patients with Parkinson’s disease may become pathologic gamblers. Other patients may develop a habit for recreational activities, such as shopping, eating compulsively, or hypersexuality. Although large-scale studies are not yet available, an estimated 1 in 7 parkinsonian patients develops an addiction-like behavior when


receiving dopamine agonists. Large individual differences exist also in vulnerability to substance-related addiction. Whereas one person may become “hooked” after a few doses, others may be able to use a drug occasionally during their entire lives without ever having difficulty in stopping. Even when dependence is induced with chronic exposure, only a small percentage of dependent users progress to addiction. Recent studies in rats suggest that impulsivity or excessive anxiety may be crucial traits that represent a risk for addiction. The transition to addiction is determined by a combination of environmental and genetic factors. Heritability of addiction, as determined by comparing monozygotic with dizygotic twins, is relatively modest for cannabinoids but very high for cocaine. It is of interest that the relative risk for addiction (addiction liability) of a drug (Table 32–1) correlates with its heritability, suggesting that the neurobiologic basis of addiction common to all drugs is what is being inherited. Further genomic analysis indicates that only a few alleles (or perhaps even a single recessive allele) need to function in combination to produce the phenotype. However, identification of the genes involved remains elusive. Although some substance-specific candidate genes have been identified (eg, alcohol dehydrogenase), future research will also focus on genes implicated in the neurobiologic mechanisms common to all addictive drugs.

NONADDICTIVE DRUGS OF ABUSE Some drugs of abuse do not lead to addiction. This is the case for substances that alter perception without causing sensations of reward and euphoria, such as the hallucinogens and the dissociative anesthetics (Table 32–1). Unlike addictive drugs, which primarily target the mesolimbic dopamine system, these agents primarily target cortical and thalamic circuits. Lysergic acid diethylamide (LSD), for example, activates the serotonin 5-HT2A receptor in the prefrontal cortex, enhancing glutamatergic transmission onto pyramidal neurons. These excitatory afferents mainly come from the thalamus and carry sensory information of varied modalities, which may constitute a link to enhanced perception. Phencyclidine (PCP) and ketamine produce a feeling of separation of mind and body (which is why they are called dissociative anesthetics) and, at higher doses, stupor and coma. The principal mechanism of action is a use-dependent inhibition of glutamate receptors of the NMDA type. High doses of dextromethorphan, an over-the-counter cough suppressant, can also elicit a dissociative state. This effect is mediated by a rather nonselective action on serotonin reuptake, and opioid, acetylcholine, and NMDA receptors. The classification of NMDA antagonists as nonaddictive drugs was based on early assessments, which, in the case of PCP, have recently been questioned. In fact, animal research shows that PCP can increase mesolimbic dopamine concentrations and has some reinforcing properties in rodents. Concurrent effects on both thalamocortical and mesolimbic systems also exist for other addictive drugs. Psychosis-like symptoms can be observed with cannabinoids, amphetamines, and cocaine, which may reflect their effects on thalamocortical structures. For example, cannabinoids, in addition to their documented effects on the mesolimbic dopamine system, also enhance excitation in cortical circuits through presynaptic inhibition of GABA release. Hallucinogens and NMDA antagonists, even if they do not produce dependence or addiction, can still have long-term effects. Flashbacks of altered perception can occur years after LSD use. Moreover, chronic use of PCP may lead to an irreversible schizophrenia-like psychosis.

BASIC PHARMACOLOGY OF DRUGS OF ABUSE Since all addictive drugs increase dopamine concentrations in target structures of the mesolimbic projections, we classify them on the basis of their molecular targets and the underlying mechanisms (Table 32–1 and Figure 32–2). The first group contains the opioids, cannabinoids, γ-hydroxybutyric acid (GHB), and the hallucinogens, which all exert their action through Gio protein-coupled receptors. The second group includes nicotine, alcohol, the benzodiazepines, dissociative anesthetics, and some inhalants, which interact with ionotropic receptors or ion channels. The last group comprises cocaine, amphetamines, and ecstasy, which all bind to monoamine transporters. The nonaddictive drugs are classified using the same criteria.

DRUGS THAT ACTIVATE GIO-COUPLED RECEPTORS OPIOIDS Opioids may have been the first drugs to be abused (preceding stimulants), and are still among the most commonly used for nonmedical purposes.

Pharmacology & Clinical Aspects As described in Chapter 31, opioids comprise a large family of endogenous and exogenous agonists at three G protein-coupled receptors: the μ-, κ-, and δ-opioid receptors. Although all three receptors couple to inhibitory G proteins (ie, they all inhibit adenylyl cyclase), they


have distinct, sometimes even opposing effects, mainly because of the cell type-specific expression throughout the brain. In the VTA, for example, μ-opioid receptors are selectively expressed on GABA neurons (which they inhibit), whereas κ-opioid receptors are expressed on and inhibit dopamine neurons. This may explain why μ-opioid agonists cause euphoria, whereas κ agonists induce dysphoria. In line with the latter observations, the rewarding effects of morphine are absent in knockout mice lacking μ receptors but persist when either of the other opioid receptors are ablated. In the VTA, μ opioids cause an inhibition of GABAergic inhibitory interneurons, which leads eventually to a disinhibition of dopamine neurons. The most commonly abused μ opioids include morphine, heroin (diacetylmorphine, which is rapidly metabolized to morphine), codeine, and oxycodone. Meperidine abuse is common among health professionals. All of these drugs induce strong tolerance and dependence. The withdrawal syndrome may be very severe (except for codeine) and includes intense dysphoria, nausea or vomiting, muscle aches, lacrimation, rhinorrhea, mydriasis, piloerection, sweating, diarrhea, yawning, and fever. Beyond the withdrawal syndrome, which usually lasts no longer than a few days, individuals who have received opioids as analgesics only rarely develop addiction. In contrast, when taken for recreational purposes, opioids are highly addictive. The relative risk of addiction is 4 out of 5 on a scale of 1 = nonaddictive, 5 = highly addictive.

Treatment The opioid antagonist naloxone reverses the effects of a dose of morphine or heroin within minutes. This may be life-saving in the case of a massive overdose (see Chapters 31 and 58). Naloxone administration also provokes an acute withdrawal (precipitated abstinence) syndrome in a dependent person who has recently taken an opioid. In the treatment of opioid addiction, a long-acting opioid (eg, methadone, buprenorphine) is often substituted for the shorter-acting, more rewarding, opioid (eg, heroin). For substitution therapy, methadone is given orally once daily, facilitating supervised intake. Using a partial agonist (buprenorphine) and the much longer half-life (methadone and buprenorphine) may also have some beneficial effects (eg, weaker drug sensitization, which typically requires intermittent exposures), but it is important to realize that abrupt termination of methadone administration invariably precipitates a withdrawal syndrome; that is, the subject on substitution therapy remains dependent. Some countries (eg, Switzerland, Netherlands) even allow substitution of heroin by heroin. A follow-up of a cohort of addicts who receive heroin injections in a controlled setting and have access to counseling indicates that addicts under heroin substitution have an improved health status and are better integrated in society.

CANNABINOIDS Endogenous cannabinoids that act as neurotransmitters include 2-arachidonyl glycerol (2-AG) and anandamide, both of which bind to CB1 receptors. These very lipid-soluble compounds are released at the postsynaptic somatodendritic membrane, and diffuse through the extracellular space to bind at presynaptic CB1 receptors, where they inhibit the release of either glutamate or GABA. Because of such backward signaling, endocannabinoids are called retrograde messengers. In the hippocampus, release of endocannabinoids from pyramidal neurons selectively affects inhibitory transmission and may contribute to the induction of synaptic plasticity during learning and memory formation. 9

Exogenous cannabinoids, eg in marijuana, include several pharmacologically active substances including Δ -tetra-hydrocannabinol (THC), a powerful psychoactive substance. Like opioids, THC causes disinhibition of dopamine neurons, mainly by presynaptic inhibition of GABA neurons in the VTA. The half-life of THC is about 4 hours. The onset of effects of THC after smoking marijuana occurs within minutes and reaches a maximum after 1–2 hours. The most prominent effects are euphoria and relaxation. Users also report feelings of well-being, grandiosity, and altered perception of passage of time. Dose-dependent perceptual changes (eg, visual distortions), drowsiness, diminished coordination, and memory impairment may occur. Cannabinoids can also create a dysphoric state and, in rare cases following the use of very high doses, eg, in hashish, result in visual hallucinations, depersonalization, and frank psychotic episodes. Additional effects of THC, eg, increased appetite, attenuation of nausea, decreased intraocular pressure, and relief of chronic pain, have led to the use of cannabinoids in medical therapeutics. The justification of medicinal use of marijuana was comprehensively examined by the Institute of Medicine (IOM) of the National Academy of Sciences in its 1999 report, Marijuana & Medicine. This continues to be a controversial issue, mainly because of the fear that cannabinoids may serve as a gateway to the consumption of “hard” drugs or cause schizophrenia in individuals with a predisposition. Chronic exposure to marijuana leads to dependence, which is revealed by a distinctive, but mild and short-lived, withdrawal syndrome that includes restlessness, irritability, mild agitation, insomnia, nausea, and cramping. The relative risk for addiction is 2. The synthetic Δ9 -THC analog dronabinol is an FDA-approved cannabinoid agonist currently marketed in the USA and some European countries. Nabilone, an older commercial Δ9 -THC analog, was recently reintroduced in the USA for treatment of chemotherapy-induced emesis. The cannabinoid system is likely to emerge as an important drug target in the future because of its apparent involvement in several therapeutically desirable effects.


GAMMA-HYDROXYBUTYRIC ACID Gamma-hydroxybutyric acid (GHB, or sodium oxybate for its salt form) is produced during the metabolism of GABA, but the function of this endogenous agent is unknown at present. The pharmacology of GHB is complex because there are two distinct binding sites. The protein that contains a high-affinity binding site (1 μM) for GHB has been cloned, but its involvement in the cellular effects of GHB at pharmacologic concentrations remains unclear. The low-affinity binding site (1 mM) has been identified as the GABA B receptor. In mice that lack GABAB receptors, even very high doses of GHB have no effect; this suggests that GABAB receptors are the sole mediators of GHB’s pharmacologic action. GHB was first synthesized in 1960 and introduced as a general anesthetic. Because of its narrow safety margin and its addictive potential, it is not available in the USA for this purpose. Sodium oxybate can, however, be prescribed (under restricted access rules) to treat narcolepsy, because GHB decreases daytime sleepiness and episodes of cataplexy through a mechanism unrelated to the reward system. Before causing sedation and coma, GHB causes euphoria, enhanced sensory perceptions, a feeling of social closeness, and amnesia. These properties have made it a popular “club drug” that goes by colorful street names such as “liquid ecstasy,” “grievous bodily harm,” or “date rape drug.” As the latter name suggests, GHB has been used in date rapes because it is odorless and can be readily dissolved in beverages. It is rapidly absorbed after ingestion and reaches a maximal plasma concentration 20–30 minutes after ingestion of a 10–20 mg/kg dose. The elimination half-life is about 30 minutes. Although GABAB receptors are expressed on all neurons of the VTA, GABA neurons are much more sensitive to GHB than are dopamine neurons (Figure 32–3). This is reflected by the EC50 s, which differ by about one order of magnitude, and indicates the difference in coupling efficiency of the GABAB receptor and the potassium channels responsible for the hyperpolarization. Because GHB is a weak agonist, only GABA neurons are inhibited at the concentrations typically obtained with recreational use. This feature may underlie the reinforcing effects of GHB and the basis for addiction to the drug. At higher doses, however, GHB also hyperpolarizes dopamine neurons, eventually completely inhibiting dopamine release. Such an inhibition of the VTA may in turn preclude its activation by other addictive drugs and may explain why GHB might have some usefulness as an “anticraving” compound.


FIGURE 32–3 Disinhibition of dopamine (DA) neurons in the ventral tegmental area (VTA) through drugs that act via Gio -coupled receptors. Top: Opioids target μ-opioid receptors (MORs) that in the VTA are located exclusively on γ-aminobutyric acid (GABA) neurons. MORs are expressed on the presynaptic terminal of these cells and the somatodendritic compartment of the postsynaptic cells. Each compartment has distinct effectors (insets). G protein-bγ-mediated inhibition of voltage-gated calcium channels (VGCC) is the major mechanism in the presynaptic terminal. Conversely, in dendrites MORs activate K channels. Middle: Δ9 -tetrahydrocannabinol (THC) and other cannabinoids mainly act through presynaptic inhibition. Bottom: Gama-hydroxybutyric acid (GHB) targets GABAB receptors, which are located on both cell types. However, GABA neurons are more sensitive to GHB than are DA neurons, leading to disinhibition at concentrations typically obtained with recreational use. CB1 R, cannabinoid receptors.

LSD, MESCALINE, & PSILOCYBIN LSD, mescaline, and psilocybin are commonly called hallucinogens because of their ability to alter consciousness such that the individual senses things that are not present. They induce, often in an unpredictable way, perceptual symptoms, including shape and color distortion.


Psychosis-like manifestations (depersonalization, hallucinations, distorted time perception) have led some to classify these drugs as psychotomimetics. They also produce somatic symptoms (dizziness, nausea, paresthesias, and blurred vision). Some users have reported intense reexperiencing of perceptual effects (flashbacks) up to several years after the last drug exposure. Hallucinogens differ from most other drugs described in this chapter in that they induce neither dependence nor addiction. However, repetitive exposure still leads to rapid tolerance (also called tachyphylaxis). Animals do not self-administer hallucinogens, suggesting that they are not rewarding to them. Additional studies show that these drugs also fail to stimulate dopamine release, further supporting the idea that only drugs that activate the mesolimbic dopamine system are addictive. Instead, hallucinogens increase glutamate release in the cortex, presumably by enhancing excitatory afferent input via presynaptic serotonin receptors (eg, 5HT2A) from the thalamus. LSD is an ergot alkaloid. After synthesis, blotter paper or sugar cubes are sprinkled with the liquid and allowed to dry. When LSD is swallowed, psychoactive effects typically appear after 30 minutes and last 6–12 hours. During this time, subjects have impaired ability to make rational judgments and understand common dangers, which puts them at risk for accidents and personal injury. In an adult, a typical dose is 20–30 mcg. LSD is not considered neurotoxic, but like most ergot alkaloids, may lead to strong contractions of the uterus that can induce abortion (see Chapter 16). The main molecular target of LSD and other hallucinogens is the 5-HT2A receptor. This receptor couples to G proteins of the Gq type and generates inositol trisphosphate (IP 3 ), leading to a release of intracellular calcium. Although hallucinogens, and LSD in particular, have been proposed for several therapeutic indications, efficacy has never been demonstrated.

DRUGS THAT MEDIATE THEIR EFFECTS VIA IONOTROPIC RECEPTORS NICOTINE In terms of numbers affected, addiction to nicotine exceeds all other forms of addiction, affecting more than 50% of all adults in some countries. Nicotine exposure occurs primarily through smoking of tobacco, which causes associated diseases that are responsible for many preventable deaths. The chronic use of chewing tobacco and snuff tobacco is also addictive. Nicotine is a selective agonist of the nicotinic acetylcholine receptor (nAChR) that is normally activated by acetylcholine (see Chapters 6 and 7). Based on nicotine’s enhancement of cognitive performance and the association of Alzheimer’s dementia with a loss of ACh-releasing neurons from the nucleus basalis of Meynert, nAChRs are believed to play an important role in many cognitive processes. The rewarding effect of nicotine requires involvement of the VTA, in which nAChRs are expressed on dopamine neurons. When nicotine excites projection neurons, dopamine is released in the nucleus accumbens and the prefrontal cortex, thus fulfilling the dopamine requirement of addictive drugs. Recent work has identified α4b2-containing channels in the VTA as the nAChRs that are required for the rewarding effects of nicotine. This statement is based on the observation that knockout mice deficient for the b2 subunit lose interest in self-administering nicotine, and that in these mice, this behavior can be restored through an in vivo transfection of the b2 subunit in neurons of the VTA. Electrophysiologic evidence suggests that homomeric nAChRs made exclusively of α7 subunits also contribute to the reinforcing effects of nicotine. These receptors are mainly expressed on synaptic terminals of excitatory afferents projecting onto the dopamine neurons. They also contribute to nicotine-evoked dopamine release and the long-term changes induced by the drugs related to addiction (eg, long-term synaptic potentiation of excitatory inputs). Nicotine withdrawal is mild compared with opioid withdrawal and involves irritability and sleep problems. However, nicotine is among the most addictive drugs (relative risk 4), and relapse after attempted cessation is very common.

Treatment Treatments for nicotine addiction include nicotine itself in forms that are slowly absorbed and several other drugs. Nicotine that is chewed, inhaled, or transdermally delivered can be substituted for the nicotine in cigarettes, thus slowing the pharmacokinetics and eliminating the many complications associated with the toxic substances found in tobacco smoke. Recently, two partial agonists of α4b2containing nAChRs have been characterized: the plant-extract cytisine and its synthetic derivative varenicline. Both work by occupying nAChRs on dopamine neurons of the VTA, thus preventing nicotine from exerting its action. Varenicline may impair the capacity to drive and has been associated with suicidal ideation. The antidepressant bupropion is approved for nicotine cessation therapy. It is most effective when combined with behavioral therapies. Many countries have banned smoking in public places to create smoke-free environments. This important step not only reduces passive smoking and the hazards of secondhand smoke, but also the risk that ex-smokers will be exposed to smoke, which as a contextual cue, may trigger relapse.

BENZODIAZEPINES Benzodiazepines are commonly prescribed as anxiolytics and sleep medications. They represent a definite risk for abuse, which has to be weighed against their beneficial effects. Benzodiazepines are abused by some persons for their euphoriant effects, but most often abuse


occurs concomitant with other drugs, eg, to attenuate anxiety during withdrawal from opioids. Barbiturates, which preceded benzodiazepines as the most commonly abused sedative-hypnotics (after ethanol), are now rarely prescribed to outpatients and therefore constitute a less common prescription drug problem than they did in the past. Street sales of barbiturates, however, continue. Management of barbiturate withdrawal and addiction is similar to that of benzodiazepines. Benzodiazepine dependence is very common, and diagnosis of addiction probably often missed. Withdrawal from benzodiazepines occurs within days of stopping the medication and varies as a function of the half-life of elimination. Symptoms include irritability, insomnia, phonophobia and photophobia, depression, muscle cramps, and even seizures. Typically, these symptoms taper off within 1–2 weeks. Benzodiazepines are positive modulators of the GABAA receptor, increasing both single-channel conductance and open-channel probability. GABA A receptors are pentameric structures consisting of α, b, and γ subunits (see Chapter 22). GABA receptors on dopamine neurons of the VTA lack α1, a subunit isoform that is present in GABA neurons nearby (ie, interneurons). Because of this difference, unitary synaptic currents in interneurons are larger than those in dopamine neurons, and when this difference is amplified by benzodiazepines, interneurons fall silent. GABA is no longer released, and benzodiazepines lose their effect on dopamine neurons, ultimately leading to disinhibition of the dopamine neurons. The rewarding effects of benzodiazepines are, therefore, mediated by α1containing GABAA receptors expressed on VTA neurons. Receptors containing α5 subunits seem to be required for tolerance to the sedative effects of benzodiazepines, and studies in humans link α2b3-containing receptors to alcohol dependence (the GABAA receptor is also a target of alcohol, see following text). Taken together, a picture is emerging linking GABA A receptors that contain the α1 subunit isoform to their addiction liability. By extension, α1-sparing compounds, which at present remain experimental and are not approved for human use, may eventually be preferred to treat anxiety disorders because of their reduced risk of induced addiction.

ALCOHOL Alcohol (ethanol, see Chapter 23) is regularly used by a majority of the population in many Western countries. Although only a minority becomes dependent and addicted, abuse is a very serious public health problem because of the social costs and many diseases associated with alcoholism.

Pharmacology The pharmacology of alcohol is complex, and no single receptor mediates all of its effects. On the contrary, alcohol alters the function of several receptors and cellular functions, including GABAA receptors, Kir3/GIRK channels, adenosine reuptake (through the equilibrative nucleoside transporter, ENT1), glycine receptor, NMDA receptor, and 5-HT 3 receptor. They are all, with the exception of ENT1, either ionotropic receptors or ion channels. It is not clear which of these targets is responsible for the increase of dopamine release from the mesolimbic reward system. The inhibition of ENT1 is probably not responsible for the rewarding effects (ENT1 knockout mice drink more than controls) but seems to be involved in alcohol dependence through an accumulation of adenosine, stimulation of adenosine A 2 receptors, and ensuing enhanced CREB signaling. Dependence becomes apparent 6–12 hours after cessation of heavy drinking as a withdrawal syndrome that may include tremor (mainly of the hands), nausea and vomiting, excessive sweating, agitation, and anxiety. In some individuals, this is followed by visual, tactile, and auditory hallucinations 12–24 hours after cessation. Generalized seizures may manifest after 24–48 hours. Finally, 48–72 hours after cessation, an alcohol withdrawal delirium (delirium tremens) may become apparent in which the person hallucinates, is disoriented, and shows evidence of autonomic instability. Delirium tremens is associated with 5–15% mortality.

Treatment Treatment of ethanol withdrawal is supportive and relies on benzodiazepines, taking care to use compounds such as oxazepam and lorazepam, which are not as dependent on oxidative hepatic metabolism as most other benzodiazepines. In patients in whom monitoring is not reliable and liver function is adequate, a longer-acting benzodiazepine such as chlordiazepoxide is preferred. As in the treatment of all chronic drug abuse problems, heavy reliance is placed on psychosocial approaches to alcohol addiction. This is perhaps even more important for the alcoholic patient because of the ubiquitous presence of alcohol in many social contexts. The pharmacologic treatment of alcohol addiction is limited, although several compounds, with different goals, have been used. Therapy is discussed in Chapter 23.

KETAMINE & PHENCYCLIDINE (PCP) Ketamine and PCP were developed as general anesthetics (see Chapter 25), but only ketamine is still used for this application. Both drugs, along with others, are now classified as “club drugs” and sold under names such as “angel dust,” “Hog,” and “Special K.” They


owe their effects to their use-dependent, noncompetitive antagonism of the NMDA receptor. The effects of these substances became apparent when patients undergoing surgery reported unpleasant vivid dreams and hallucinations after anesthesia. Ketamine and PCP are white crystalline powders in their pure forms, but on the street they are also sold as liquids, capsules, or pills, which can be snorted, ingested, injected, or smoked. Psychedelic effects last for about 1 hour and also include increased blood pressure, impaired memory function, and visual alterations. At high doses, unpleasant out-of-body and near-death experiences have been reported. Although ketamine and phencyclidine do not cause dependence and addiction (relative risk = 1), chronic exposure, particularly to PCP, may lead to long-lasting psychosis closely resembling schizophrenia, which may persist beyond drug exposure. Surprisingly, intravenous administration of ketamine can eliminate episodes of depression within hours (see Chapter 30), which is in strong contrast to selective serotonin reuptake inhibitors and other antidepressants, which usually take weeks to act. The antidepressive mechanism is believed to involve the antagonism of NMDA receptors, thus favoring the mTOR pathway downstream of other glutamate receptors. A limitation of this approach is the transient nature of the effect, which wears off within days even with repetitive administration.

INHALANTS Inhalant abuse is defined as recreational exposure to chemical vapors, such as nitrates, ketones, and aliphatic and aromatic hydrocarbons. These substances are present in a variety of household and industrial products that are inhaled by “sniffing,” “huffing,” or “bagging.” Sniffing refers to inhalation from an open container, huffing to the soaking of a cloth in the volatile substance before inhalation, and bagging to breathing in and out of a paper or plastic bag filled with fumes. It is common for novices to start with sniffing and progress to huffing and bagging as addiction develops. Inhalant abuse is particularly prevalent in children and young adults. The exact mechanism of action of most volatile substances remains unknown. Altered function of ionotropic receptors and ion channels throughout the central nervous system has been demonstrated for a few. Nitrous oxide, for example, binds to NMDA receptors and fuel additives enhance GABAA receptor function. Most inhalants produce euphoria; increased excitability of the VTA has been documented for toluene and may underlie its addiction risk. Other substances, such as amyl nitrite (“poppers”), primarily produce smooth muscle relaxation and enhance erection, but are not addictive. With chronic exposure to the aromatic hydrocarbons (eg, benzene, toluene), toxic effects can be observed in many organs, including white matter lesions in the central nervous system. Management of overdose remains supportive.

DRUGS THAT BIND TO TRANSPORTERS OF BIOGENIC AMINES Cocaine The prevalence of cocaine abuse has increased greatly over the last decade and now represents a major public health problem worldwide. Cocaine is highly addictive (relative risk = 5), and its use is associated with a number of complications. Cocaine is an alkaloid found in the leaves of Erythroxylon coca, a shrub indigenous to the Andes. For more than 100 years, it has been extracted and used in clinical medicine, mainly as a local anesthetic and to dilate pupils in ophthalmology. Sigmund Freud famously proposed its use to treat depression and alcohol dependence, but addiction quickly brought an end to this idea. Cocaine hydrochloride is a water-soluble salt that can be injected or absorbed by any mucosal membrane (eg, nasal snorting). When heated in an alkaline solution, it is transformed into the free base, “crack cocaine,” which can then be smoked. Inhaled crack cocaine is rapidly absorbed in the lungs and penetrates swiftly into the brain, producing an almost instantaneous “rush.” In the peripheral nervous system, cocaine inhibits voltage-gated sodium channels, thus blocking initiation and conduction of action potentials (see Chapter 26). This effect, however, seems responsible for neither the acute rewarding nor the addictive effects. In the central nervous system, cocaine blocks the uptake of dopamine, noradrenaline, and serotonin through their respective transporters. The block of the dopamine transporter (DAT), by increasing dopamine concentrations in the nucleus accumbens, has been implicated in the rewarding effects of cocaine (Figure 32–4). In fact, the rewarding effects of cocaine are abolished in mice with a cocaine-insensitive DAT. The activation of the sympathetic nervous system results mainly from blockage of the norepinephrine transporter (NET) and leads to an acute increase in arterial pressure, tachycardia, and often, ventricular arrhythmias. Users typically lose their appetite, are hyperactive, and sleep little. Cocaine exposure increases the risk for intracranial hemorrhage, ischemic stroke, myocardial infarction, and seizures. Cocaine overdose may lead to hyperthermia, coma, and death. In the 1970s, when crack-cocaine appeared in the USA, it was suggested that the drug is particularly harmful to the fetus in addicted pregnant women. The term “crack-baby” was used to describe a specific syndrome of the newborn, and the mothers faced harsh legal consequences. The follow-up of the children, now adults, does not confirm a drug-specific handicap in cognitive performance. Moreover, in this population the percentage of drug-users is comparable to controls matched for socioeconomic environment.


FIGURE 32–4 Mechanism of action of cocaine and amphetamine on synaptic terminal of dopamine (DA) neurons. Left: Cocaine inhibits the dopamine transporter (DAT), decreasing DA clearance from the synaptic cleft and causing an increase in extracellular DA concentration. Right: Since amphetamine (Amph) is a substrate of the DAT, it competitively inhibits DA transport. In addition, once in the cell, amphetamine interferes with the vesicular monoamine transporter (VMAT) and impedes the filling of synaptic vesicles. As a consequence, vesicles are depleted and cytoplasmic DA increases. This leads to a reversal of DAT direction, strongly increasing nonvesicular release of DA, and further increasing extracellular DA concentrations. Susceptible individuals may become dependent and addicted after only a few exposures to cocaine. Although a withdrawal syndrome is reported, it is not as strong as that observed with opioids. Tolerance may develop, but in some users a reverse tolerance is observed; that is, they become sensitized to small doses of cocaine. This behavioral sensitization is in part context-dependent. Cravings are very strong and underline the very high addiction liability of cocaine. To date, no specific antagonist is available, and the management of intoxication remains supportive. Developing a pharmacologic treatment for cocaine addiction is a top priority.

AMPHETAMINES Amphetamines are a group of synthetic, indirect-acting sympathomimetic drugs that cause the release of endogenous biogenic amines, such as dopamine and noradrenaline (see Chapters 6 and 9). Amphetamine, methamphetamine, and their many derivatives exert their effects by reversing the action of biogenic amine transporters at the plasma membrane. Amphetamines are substrates of these transporters and are taken up into the cell (Figure 32–4). Once in the cell, amphetamines interfere with the vesicular monoamine transporter (VMAT; see Figure 6–4), depleting synaptic vesicles of their neurotransmitter content. As a consequence, levels of dopamine (or other transmitter amine) in the cytoplasm increase and quickly become sufficient to cause release into the synapse by reversal of the plasma membrane DAT. Normal vesicular release of dopamine consequently decreases (because synaptic vesicles contain less transmitter), whereas nonvesicular release increases. Similar mechanisms apply for other biogenic amines (serotonin and norepinephrine). Together with GHB and ecstasy, amphetamines are often referred to as “club drugs” because they are increasingly popular in the club scene. They are often produced in small clandestine laboratories, which makes their precise chemical identification difficult. They differ from ecstasy chiefly in the context of use: intravenous administration and “hard-core” addiction is far more common with amphetamines, especially methamphetamine. In general, amphetamines lead to elevated catecholamine levels that increase arousal and reduce sleep, whereas the effects on the dopamine system mediate euphoria but may also cause abnormal movements and precipitate psychotic episodes. Effects on serotonin transmission may play a role in the hallucinogenic and anorexigenic functions as well as in the hyperthermia often caused by amphetamines. Unlike many other abused drugs, amphetamines are neurotoxic. The exact mechanism is not known, but neurotoxicity depends on the NMDA receptor and affects mainly serotonin and dopamine neurons. Amphetamines are typically taken initially in pill form by abusers, but can also be smoked or injected. Heavy users often progress rapidly to intravenous administration. Within hours after oral ingestion, amphetamines increase alertness and cause euphoria, agitation, and confusion. Bruxism (tooth grinding) and skin flushing may occur. Effects on heart rate may be minimal with some compounds (eg, methamphetamine), but with increasing dosage these agents often lead to tachycardia and dysrhythmias. Hypertensive crisis and


vasoconstriction may lead to stroke. Spread of HIV and hepatitis infection in inner cities has been closely associated with needle sharing by intravenous users of methamphetamine. With chronic use, amphetamine tolerance may develop, leading to dose escalation. Withdrawal consists of dysphoria, drowsiness (in some cases, insomnia), and general irritability.

ECSTASY (MDMA) Ecstasy is the name of a class of drugs that includes a large variety of derivatives of the amphetamine-related compound methylenedioxymethamphetamine (MDMA). MDMA was originally used in some forms of psychotherapy, but no medically useful effects were documented. This is perhaps not surprising, because the main effect of ecstasy appears to be to foster feelings of intimacy and empathy without impairing intellectual capacities. Today, MDMA and its many derivatives are often produced in small quantities in ad hoc laboratories and distributed at parties or “raves,” where it is taken orally. Ecstasy therefore is the prototypic designer drug and, as such, is increasingly popular. Similar to the amphetamines, MDMA causes release of biogenic amines by reversing the action of their respective transporters. It has a preferential affinity for the serotonin transporter (SERT) and therefore most strongly increases the extracellular concentration of serotonin. This release is so profound that there is a marked intracellular depletion for 24 hours after a single dose. With repetitive administration, serotonin depletion may become permanent, which has triggered a debate on its neurotoxicity. Although direct proof from animal models for neurotoxicity remains weak, several studies report long-term cognitive impairment in heavy users of MDMA. In contrast, there is a wide consensus that MDMA has several acute toxic effects, in particular hyperthermia, which along with dehydration (eg, caused by an all-night dance party) may be fatal. Other complications include serotonin syndrome (mental status change, autonomic hyperactivity, and neuromuscular abnormalities, see Chapter 16) and seizures. Following warnings about the dangers of MDMA, some users have attempted to compensate for hyperthermia by drinking excessive amounts of water, causing water intoxication involving severe hyponatremia, seizures, and even death. Withdrawal is marked by a mood “offset” characterized by depression lasting up to several weeks. There have also been reports of increased aggression during periods of abstinence in chronic MDMA users. Taken together, the evidence for irreversible damage to the brain, although not completely convincing, implies that even occasional recreational use of MDMA cannot be considered safe.

CLINICAL PHARMACOLOGY OF DEPENDENCE & ADDICTION To date no single pharmacologic treatment (even in combination with behavioral interventions) efficiently eliminates addiction. This is not to say that addiction is irreversible. Pharmacologic interventions may in fact be useful at all stages of the disease. This is particularly true in the case of a massive overdose, in which reversal of drug action may be a life-saving measure. However, in this regard, FDAapproved antagonists are available only for opioids and benzodiazepines. Pharmacologic interventions may also aim to alleviate the withdrawal syndrome, particularly after opioid exposure. On the assumption that withdrawal reflects at least in part a hyperactivity of central adrenergic systems, the α2 -adrenoceptor agonist clonidine (also used as a centrally active antihypertensive drug, see Chapter 11) has been used with some success to attenuate withdrawal. Today, most clinicians prefer to manage opioid withdrawal by very slowly tapering the administration of long-acting opioids. Another widely accepted treatment is substitution of a legally available agonist that acts at the same receptor as the abused drug. This approach has been approved for opioids and nicotine. For example, heroin addicts may receive methadone to replace heroin; smoking addicts may receive nicotine continuously via a transdermal patch system to replace smoking. In general, a rapid-acting substance is replaced with one that acts or is absorbed more slowly. Substitution treatments are largely justified by the benefits of reducing associated health risks, the reduction of drug-associated crime, and better social integration. Although dependence persists, it may be possible, with the support of behavioral interventions, to motivate drug users to gradually reduce the dose and become abstinent. The biggest challenge is the treatment of addiction itself. Several approaches have been proposed, but all remain experimental. One approach is to pharmacologically reduce cravings. The μ-opioid receptor antagonist and partial agonist naltrexone is FDA-approved for this indication in opioid and alcohol addiction. Its effect is modest and may involve a modulation of endogenous opioid systems. Clinical trials are currently being conducted with a number of drugs, including the high-affinity GABAB-receptor agonist baclofen, and initial results have shown a significant reduction of craving. This effect may be mediated by the inhibition of the dopamine neurons of the VTA, which is possible at baclofen concentrations obtained by oral administration because of its very high affinity for the GABA B receptor. Rimonabant is an inverse agonist of the CB1 receptor that behaves like an antagonist of cannabinoids. It was developed for smoking cessation and to facilitate weight loss. Because of frequent adverse effects—most notably severe depression carrying a substantial risk of suicide—this drug is no longer used clinically. It was initially used in conjunction with diet and exercise for patients with a body mass index above 30 kg/m2 (27 kg/m2 if associated risk factors, such as type 2 diabetes or dyslipidemia are present). Although a recent large-


scale study confirmed that rimonabant is effective for smoking cessation and the prevention of weight gain in smokers who quit, this indication has never been approved. While the cellular mechanism of rimonabant remains to be elucidated, data in rodents convincingly demonstrate that this compound can reduce self-administration in naive as well as drug-experienced animals.

SUMMARY Drugs Used to Treat Dependence and Addiction



REFERENCES General Goldman D, Oroszi G, Ducci F: T he genetics of addictions: Uncovering the genes. Nat Rev Genet 2005;6:521. Koob GF, Volkov ND: Neurocircuitry of addiction. Neuropsychopharmacology 2010;35:217. Lüscher C: Disease focus: Drug-evoked synaptic plasticity causing addictive behavior. J Neurosci 2013;33:17641. Lüscher C, Malenka RC: Synaptic plasticity in addiction: From molecular changes to circuit remodeling. Neuron 2011;69:650. Lüscher C, Ungless MA: T he mechanistic classification of addictive drugs. PLoS Med 2006;3:e437. Redish AD, Jensen S, Johnson A: A unified framework for addiction: Vulnerabilities in the decision process. Behav Brain Sci 2008;31:461.

Pharmacology of Drugs of Abuse Benowitz NL: Nicotine addiction. N Engl J Med 2010;362:2295. Maskos U et al: Nicotine reinforcement and cognition restored by targeted expression of nicotinic receptors. Nature 2005;436:103. Morton J: Ecstasy: Pharmacology and neurotoxicity. Curr Opin Pharmacol 2005;5:79. Nichols DE: Hallucinogens. Pharmacol T her 2004;101:131. Snead OC, Gibson KM: Gamma-hydroxybutyric acid. N Engl J Med 2005;352:2721. Sulzer D et al: Mechanisms of neurotransmitter release by amphetamines: A review. Prog Neurobiol 2005;75:406. T an KR et al: Neural basis for addictive properties of benzodiazepines. Nature 2010;463:769.

CASE STUDY ANSWER Mr V fulfills the criteria for addiction, because he has an excessive and compulsive consumption of cocaine despite the negative consequences for his job. He is certainly also alcohol dependent, and abrupt termination will likely lead to a withdrawal syndrome (eg, agitation, hallucinations, tremor, seizures, etc). His drug abuse began in late adolescence, which is usually considered a critical period. The case also illustrates how addicts use different drugs, in part to “treat” side effects (eg, cannabis or alcohol to relax after cocaine use).


SECTION VI DRUGS USED TO TREAT DISEASES OF THE BLOOD, INFLAMMATION, & GOUT


CHAPTER

33 Agents Used in Cytopenias; Hematopoietic Growth Factors James L. Zehnder, MD*

CASE STUDY A 65-year-old woman with a long-standing history of poorly controlled type 2 diabetes mellitus presents with increasing numbness and paresthesias in her extremities, generalized weakness, a sore tongue, and gastrointestinal discomfort. Physical examination reveals a frail-looking, pale woman with diminished vibration sensation, diminished spinal reflexes, and a positive Babinski sign. Examination of her oral cavity reveals atrophic glossitis, in which the tongue appears deep red in color and abnormally smooth and shiny due to atrophy of the lingual papillae. Laboratory testing reveals a macrocytic anemia based on a hematocrit of 30% (normal for women, 37–48%), a hemoglobin concentration of 9.4 g/dL (normal for elderly women, 11.7–13.8 g/dL), an erythrocyte mean cell volume (MCV) of 123 fL (normal, 84–99 fL), an erythrocyte mean cell hemoglobin concentration (MCHC) of 34% (normal, 31–36%), and a low reticulocyte count. Further laboratory testing reveals a normal serum folate concentration and a serum vitamin B12 (cobalamin) concentration of 98 pg/mL (normal, 250–1100 pg/mL). Results of a Schilling test indicate a diagnosis of pernicious anemia. Once megaloblastic anemia was identified, why was it important to measure serum concentrations of both folic acid and cobalamin? Should this patient be treated with oral or parenteral vitamin B12 ?

Hematopoiesis, the production from undifferentiated stem cells of circulating erythrocytes, platelets, and leukocytes, is a remarkable process that produces over 200 billion new blood cells per day in the normal person and even greater numbers of cells in people with conditions that cause loss or destruction of blood cells. The hematopoietic machinery resides primarily in the bone marrow in adults and requires a constant supply of three essential nutrients—iron, vitamin B 12 , and folic acid—as well as the presence of hematopoietic growth factors, proteins that regulate the proliferation and differentiation of hematopoietic cells. Inadequate supplies of either the essential nutrients or the growth factors result in deficiency of functional blood cells. Anemia, a deficiency in oxygen-carrying erythrocytes, is the most common and several forms are easily treated. Sickle cell anemia, a condition resulting from a genetic alteration in the hemoglobin molecule, is common but is not easily treated. It is discussed in the Box: Sickle Cell Disease and Hydroxyurea. Thrombocytopenia and neutropenia are not rare, and some forms are amenable to drug therapy. In this chapter, we first consider treatment of anemia due to deficiency of iron, vitamin B12 , or folic acid and then turn to the medical use of hematopoietic growth factors to combat anemia, thrombocytopenia, and neutropenia, and to support stem cell transplantation.

AGENTS USED IN ANEMIAS IRON Basic Pharmacology Iron deficiency is the most common cause of chronic anemia. Like other forms of chronic anemia, iron deficiency anemia leads to pallor, fatigue, dizziness, exertional dyspnea, and other generalized symptoms of tissue hypoxia. The cardiovascular adaptations to chronic anemia—tachycardia, increased cardiac output, vasodilation—can worsen the condition of patients with underlying cardiovascular disease. Iron forms the nucleus of the iron-porphyrin heme ring, which together with globin chains forms hemoglobin. Hemoglobin reversibly binds oxygen and provides the critical mechanism for oxygen delivery from the lungs to other tissues. In the absence of adequate iron, small erythrocytes with insufficient hemoglobin are formed, giving rise to microcytic hypochromic anemia. Iron-containing heme is


also an essential component of myoglobin, cytochromes, and other proteins with diverse biologic functions.

Pharmacokinetics Free inorganic iron is extremely toxic, but iron is required for essential proteins such as hemoglobin; therefore, evolution has provided an elaborate system for regulating iron absorption, transport, and storage (Figure 33–1). The system uses specialized transport, storage, ferrireductase, and ferroxidase proteins whose concentrations are controlled by the body’s demand for hemoglobin synthesis and adequate iron stores (Table 33–1). A peptide called hepcidin, produced primarily by liver cells, serves as a key central regulator of the system. Nearly all of the iron used to support hematopoiesis is reclaimed from catalysis of the hemoglobin in senescent or damaged erythrocytes. Normally, only a small amount of iron is lost from the body each day, so dietary requirements are small and easily fulfilled by the iron available in a wide variety of foods. However, in special populations with either increased iron requirements (eg, growing children, pregnant women) or increased losses of iron (eg, menstruating women), iron requirements can exceed normal dietary supplies, and iron deficiency can develop.


FIGURE 33–1 Absorption, transport, and storage of iron. Intestinal epithelial cells actively absorb inorganic iron via the divalent metal transporter 1 (DMT1) and heme iron via the heme carrier protein 1 (HCP1). Iron that is absorbed or released from absorbed heme iron in the intestine (1) is actively transported into the blood by ferroportin (FP) or complexed with apoferritin (AF) and stored as ferritin (F). In the blood, iron is transported by transferrin (Tf) to erythroid precursors in the bone marrow for synthesis of hemoglobin (Hgb) (2) or to hepatocytes for storage as ferritin (3). The transferrin-iron complex binds to transferrin receptors (TfR) in erythroid precursors and hepatocytes and is internalized. After release of iron, the TfR-Tf complex is recycled to the plasma membrane and Tf is released. Macrophages that phagocytize senescent erythrocytes (RBC) reclaim the iron from the RBC hemoglobin and either export it or store it as ferritin (4). Hepatocytes use several mechanisms to take up iron and store the iron as ferritin. FO, ferroxidase. (Reproduced, with permission, from Trevor A et al: Pharmacology Examination & Board Review, 9th ed. McGraw-Hill, 2010. Copyright © The McGraw-Hill Companies, Inc.) TABLE 33–1 Iron distribution in normal adults.1

Sickle Cell Disease and Hydroxyurea Sickle cell disease is an important genetic cause of hemolytic anemia, a form of anemia due to increased erythrocyte destruction, instead of the reduced mature erythrocyte production seen with iron, folic acid, and vitamin B12 deficiency. Patients with sickle cell disease are homozygous for the aberrant β-hemoglobin S (HbS) allele (substitution of valine for glutamic acid at amino acid 6 of βglobin) or heterozygous for HbS and a second mutated β-hemoglobin gene such as hemoglobin C (HbC) or β-thalassemia. Sickle cell disease has an increased prevalence in individuals of African descent because the heterozygous trait confers resistance to malaria. In the majority of patients with sickle cell disease, anemia is not the major problem; the anemia is generally well compensated even though such individuals have a chronically low hematocrit (20–30%), a low serum hemoglobin level (7–10 g/dL), and an elevated reticulocyte count. Instead, the primary problem is that deoxygenated HbS chains form polymeric structures that dramatically change erythrocyte shape, reduce deformability, and elicit membrane permeability changes that further promote hemoglobin polymerization. Abnormal erythrocytes aggregate in the microvasculature—where oxygen tension is low and hemoglobin is deoxygenated—and cause veno-occlusive damage. In the musculoskeletal system, this results in characteristic,


extremely severe bone and joint pain. In the cerebral vascular system, it causes ischemic stroke. Damage to the spleen increases the risk of infection, particularly by encapsulated bacteria such as Streptococcus pneumoniae. In the pulmonary system, there is an increased risk of infection and, in adults, an increase in embolism and pulmonary hypertension. Supportive treatment includes analgesics, antibiotics, pneumococcal vaccination, and blood transfusions. In addition, the cancer chemotherapeutic drug hydroxyurea (hydroxycarbamide) reduces veno-occlusive events. It is approved in the United States for treatment of adults with recurrent sickle cell crises and approved in Europe in adults and children with recurrent vaso-occlusive events. As an anticancer drug used in the treatment of chronic and acute myelogenous leukemia, hydroxyurea inhibits ribonucleotide reductase and thereby depletes deoxynucleoside triphosphate and arrests cells in the S phase of the cell cycle (see Chapter 54). In the treatment of sickle cell disease, hydroxyurea acts through poorly defined pathways to increase the production of fetal hemoglobin γ (HbF), which interferes with the polymerization of HbS. Clinical trials have shown that hydroxyurea decreases painful crises in adults and children with severe sickle cell disease. Its adverse effects include hematopoietic depression, gastrointestinal effects, and teratogenicity in pregnant women. A. Absorption The average American diet contains 10–15 mg of elemental iron daily. A normal individual absorbs 5–10% of this iron, or about 0.5–1 mg daily. Iron is absorbed in the duodenum and proximal jejunum, although the more distal small intestine can absorb iron if necessary. Iron absorption increases in response to low iron stores or increased iron requirements. Total iron absorption increases to 1–2 mg/d in menstruating women and may be as high as 3–4 mg/d in pregnant women. Iron is available in a wide variety of foods but is especially abundant in meat. The iron in meat protein can be efficiently absorbed, because heme iron in meat hemoglobin and myoglobin can be absorbed intact without first having to be dissociated into elemental iron (Figure 33–1). Iron in other foods, especially vegetables and grains, is often tightly bound to organic compounds and is much less available for absorption. Nonheme iron in foods and iron in inorganic iron salts and complexes must be reduced by a ferrireductase to ferrous iron (Fe2+) before it can be absorbed by intestinal mucosal cells. Iron crosses the luminal membrane of the intestinal mucosal cell by two mechanisms: active transport of ferrous iron by the divalent metal transporter DMT1, and absorption of iron complexed with heme (Figure 33–1). Together with iron split from absorbed heme, the newly absorbed iron can be actively transported into the blood across the basolateral membrane by a transporter known as ferroportin and oxidized to ferric iron (Fe3+) by the ferroxidase hephaestin. The liver-derived hepcidin inhibits intestinal cell iron release by binding to ferroportin and triggering its internalization and destruction. Excess iron is stored in intestinal epithelial cells as ferritin, a water-soluble complex consisting of a core of ferric hydroxide covered by a shell of a specialized storage protein called apoferritin. B. Transport Iron is transported in the plasma bound to transferrin, a β-globulin that can bind two molecules of ferric iron (Figure 33–1). The transferrin-iron complex enters maturing erythroid cells by a specific receptor mechanism. Transferrin receptors—integral membrane glycoproteins present in large numbers on proliferating erythroid cells—bind and internalize the transferrin-iron complex through the process of receptor-mediated endocytosis. In endosomes, the ferric iron is released, reduced to ferrous iron, and transported by DMT1 into the cytoplasm, where it is funneled into hemoglobin synthesis or stored as ferritin. The transferrin-transferrin receptor complex is recycled to the cell membrane, where the transferrin dissociates and returns to the plasma. This process provides an efficient mechanism for supplying the iron required by developing red blood cells. Increased erythropoiesis is associated with an increase in the number of transferrin receptors on developing erythroid cells and a reduction in hepatic hepcidin release. Iron store depletion and iron deficiency anemia are associated with an increased concentration of serum transferrin. C. Storage In addition to the storage of iron in intestinal mucosal cells, iron is also stored, primarily as ferritin, in macrophages in the liver, spleen, and bone, and in parenchymal liver cells (Figure 33–1). The mobilization of iron from macrophages and hepatocytes is primarily controlled by hepcidin regulation of ferroportin activity. Low hepcidin concentrations result in iron release from these storage sites; high hepcidin concentrations inhibit iron release. Ferritin is detectable in serum. Since the ferritin present in serum is in equilibrium with storage ferritin in reticuloendothelial tissues, the serum ferritin level can be used to estimate total body iron stores. D. Elimination There is no mechanism for excretion of iron. Small amounts are lost in the feces by exfoliation of intestinal mucosal cells, and trace amounts are excreted in bile, urine, and sweat. These losses account for no more than 1 mg of iron per day. Because the body’s ability to excrete iron is so limited, regulation of iron balance must be achieved by changing intestinal absorption and storage of iron in response to the body’s needs. As noted below, impaired regulation of iron absorption leads to serious pathology.


Clinical Pharmacology A. Indications for the Use of Iron The only clinical indication for the use of iron preparations is the treatment or prevention of iron deficiency anemia. This manifests as a hypochromic, microcytic anemia in which the erythrocyte mean cell volume (MCV) and the mean cell hemoglobin concentration are low (Table 33–2). Iron deficiency is commonly seen in populations with increased iron requirements. These include infants, especially premature infants; children during rapid growth periods; pregnant and lactating women; and patients with chronic kidney disease who lose erythrocytes at a relatively high rate during hemodialysis and also form them at a high rate as a result of treatment with the erythrocyte growth factor erythropoietin (see below). Inadequate iron absorption can also cause iron deficiency. This is seen after gastrectomy and in patients with severe small bowel disease that results in generalized malabsorption. TABLE 33–2 Distinguishing features of the nutritional anemias.

The most common cause of iron deficiency in adults is blood loss. Menstruating women lose about 30 mg of iron with each menstrual period; women with heavy menstrual bleeding may lose much more. Thus, many premenopausal women have low iron stores or even iron deficiency. In men and postmenopausal women, the most common site of blood loss is the gastrointestinal tract. Patients with


unexplained iron deficiency anemia should be evaluated for occult gastrointestinal bleeding. B. Treatment Iron deficiency anemia is treated with oral or parenteral iron preparations. Oral iron corrects the anemia just as rapidly and completely as parenteral iron in most cases if iron absorption from the gastrointestinal tract is normal. An exception is the high requirement for iron of patients with advanced chronic kidney disease who are undergoing hemodialysis and treatment with erythropoietin; for these patients, parenteral iron administration is preferred. 1. Oral iron therapy—A wide variety of oral iron preparations is available. Because ferrous iron is most efficiently absorbed, ferrous salts should be used. Ferrous sulfate, ferrous gluconate, and ferrous fumarate are all effective and inexpensive and are recommended for the treatment of most patients. Different iron salts provide different amounts of elemental iron, as shown in Table 33–3. In an iron-deficient individual, about 50–100 mg of iron can be incorporated into hemoglobin daily, and about 25% of oral iron given as ferrous salt can be absorbed. Therefore, 200– 400 mg of elemental iron should be given daily to correct iron deficiency most rapidly. Patients unable to tolerate such large doses of iron can be given lower daily doses of iron, which results in slower but still complete correction of iron deficiency. Treatment with oral iron should be continued for 3–6 months after correction of the cause of the iron loss. This corrects the anemia and replenishes iron stores. TABLE 33–3 Some commonly used oral iron preparations.

Common adverse effects of oral iron therapy include nausea, epigastric discomfort, abdominal cramps, constipation, and diarrhea. These effects are usually dose-related and can often be overcome by lowering the daily dose of iron or by taking the tablets immediately after or with meals. Some patients have less severe gastrointestinal adverse effects with one iron salt than another and benefit from changing preparations. Patients taking oral iron develop black stools; this has no clinical significance in itself but may obscure the diagnosis of continued gastrointestinal blood loss. 2. Parenteral iron therapy—Parenteral therapy should be reserved for patients with documented iron deficiency who are unable to tolerate or absorb oral iron and for patients with extensive chronic anemia who cannot be maintained with oral iron alone. This includes patients with advanced chronic renal disease requiring hemodialysis and treatment with erythropoietin, various postgastrectomy conditions and previous small bowel resection, inflammatory bowel disease involving the proximal small bowel, and malabsorption syndromes. The challenge with parenteral iron therapy is that parenteral administration of inorganic free ferric iron produces serious dosedependent toxicity, which severely limits the dose that can be administered. However, when the ferric iron is formulated as a colloid containing particles with a core of iron oxyhydroxide surrounded by a core of carbohydrate, bioactive iron is released slowly from the stable colloid particles. In the United States, the three traditional forms of parenteral iron are iron dextran, sodium ferric gluconate complex, and iron sucrose. Two newer preparations are available (see below). Iron dextran is a stable complex of ferric oxyhydroxide and dextran polymers containing 50 mg of elemental iron per milliliter of


solution. It can be given by deep intramuscular injection or by intravenous infusion, although the intravenous route is used most commonly. Intravenous administration eliminates the local pain and tissue staining that often occur with the intramuscular route and allows delivery of the entire dose of iron necessary to correct the iron deficiency at one time. Adverse effects of intravenous iron dextran therapy include headache, light-headedness, fever, arthralgias, nausea and vomiting, back pain, flushing, urticaria, bronchospasm, and, rarely, anaphylaxis and death. Owing to the risk of a hypersensitivity reaction, a small test dose of iron dextran should always be given before full intramuscular or intravenous doses are given. Patients with a strong history of allergy and patients who have previously received parenteral iron dextran are more likely to have hypersensitivity reactions after treatment with parenteral iron dextran. The iron dextran formulations used clinically are distinguishable as high-molecular-weight and low-molecular-weight forms. In the United States, the InFeD preparation is a low-molecular-weight form while DexFerrum is a high-molecular-weight form. Clinical data—primarily from observational studies—indicate that the risk of anaphylaxis is largely associated with high-molecular-weight formulations. Sodium ferric gluconate complex and iron-sucrose complex are alternative parenteral iron preparations. Ferric carboxymaltose is a colloidal iron preparation embedded within a carbohydrate polymer. Ferumoxytol is a superparamagnetic iron oxide nanoparticle coated with carbohydrate. The carbohydrate shell is removed in the reticuloendothelial system, allowing the iron to be stored as ferritin, or released to transferrin. Ferumoxytol may interfere with magnetic resonance imaging (MRI) studies. Thus if imaging is needed, MRI should be performed prior to ferumoxytol therapy or alternative imaging modality used if needed soon after dosing. These agents can be given only by the intravenous route. They appear to be less likely than high-molecular-weight iron dextran to cause hypersensitivity reactions. For patients treated chronically with parenteral iron, it is important to monitor iron storage levels to avoid the serious toxicity associated with iron overload. Unlike oral iron therapy, which is subject to the regulatory mechanism provided by the intestinal uptake system, parenteral administration—which bypasses this regulatory system—can deliver more iron than can be safely stored. Iron stores can be estimated on the basis of serum concentrations of ferritin and the transferrin saturation, which is the ratio of the total serum iron concentration to the total iron-binding capacity (TIBC).

Clinical Toxicity A. Acute Iron Toxicity Acute iron toxicity is seen almost exclusively in young children who accidentally ingest iron tablets. As few as 10 tablets of any of the commonly available oral iron preparations can be lethal in young children. Adult patients taking oral iron preparations should be instructed to store tablets in child-proof containers out of the reach of children. Children who are poisoned with oral iron experience necrotizing gastroenteritis with vomiting, abdominal pain, and bloody diarrhea followed by shock, lethargy, and dyspnea. Subsequently, improvement is often noted, but this may be followed by severe metabolic acidosis, coma, and death. Urgent treatment is necessary. Whole bowel irrigation (see Chapter 58) should be performed to flush out unabsorbed pills. Deferoxamine, a potent iron-chelating compound, can be given intravenously to bind iron that has already been absorbed and to promote its excretion in urine and feces. Activated charcoal, a highly effective adsorbent for most toxins, does not bind iron and thus is ineffective. Appropriate supportive therapy for gastrointestinal bleeding, metabolic acidosis, and shock must also be provided. B. Chronic Iron Toxicity Chronic iron toxicity (iron overload), also known as hemochromatosis, results when excess iron is deposited in the heart, liver, pancreas, and other organs. It can lead to organ failure and death. It most commonly occurs in patients with inherited hemochromatosis, a disorder characterized by excessive iron absorption, and in patients who receive many red cell transfusions over a long period of time (eg, individuals with β-thalassemia). Chronic iron overload in the absence of anemia is most efficiently treated by intermittent phlebotomy. One unit of blood can be removed every week or so until all of the excess iron is removed. Iron chelation therapy using parenteral deferoxamine or the oral iron chelator deferasirox (see Chapter 57) is less efficient as well as more complicated, expensive, and hazardous, but it may be the only option for iron overload that cannot be managed by phlebotomy, as is the case for many individuals with inherited and acquired causes of refractory anemia such as thalassemia major, sickle cell anemia, aplastic anemia, etc.

VITAMIN B 12 Vitamin B12 (cobalamin) serves as a cofactor for several essential biochemical reactions in humans. Deficiency of vitamin B12 leads to megaloblastic anemia (Table 33–2), gastrointestinal symptoms, and neurologic abnormalities. Although deficiency of vitamin B 12 due to an inadequate supply in the diet is unusual, deficiency of B12 in adults—especially older adults—due to inadequate absorption of dietary vitamin B12 is a relatively common and easily treated disorder.


Chemistry Vitamin B 12 consists of a porphyrin-like ring with a central cobalt atom attached to a nucleotide. Various organic groups may be covalently bound to the cobalt atom, forming different cobalamins. Deoxyadenosylcobalamin and methylcobalamin are the active forms of the vitamin in humans. Cyanocobalamin and hydroxocobalamin (both available for therapeutic use) and other cobalamins found in food sources are converted to the active forms. The ultimate source of vitamin B12 is from microbial synthesis; the vitamin is not synthesized by animals or plants. The chief dietary source of vitamin B12 is microbially derived vitamin B12 in meat (especially liver), eggs, and dairy products. Vitamin B12 is sometimes called extrinsic factor to differentiate it from intrinsic factor, a protein secreted by the stomach that is required for gastrointestinal uptake of dietary vitamin B12 .

Pharmacokinetics The average American diet contains 5–30 mcg of vitamin B 12 daily, 1–5 mcg of which is usually absorbed. The vitamin is avidly stored, primarily in the liver, with an average adult having a total vitamin B12 storage pool of 3000–5000 mcg. Only trace amounts of vitamin B12 are normally lost in urine and stool. Because the normal daily requirements of vitamin B12 are only about 2 mcg, it would take about 5 years for all of the stored vitamin B12 to be exhausted and for megaloblastic anemia to develop if B12 absorption were stopped. Vitamin B12 is absorbed after it complexes with intrinsic factor, a glycoprotein secreted by the parietal cells of the gastric mucosa. Intrinsic factor combines with the vitamin B12 that is liberated from dietary sources in the stomach and duodenum, and the intrinsic factor-vitamin B12 complex is subsequently absorbed in the distal ileum by a highly selective receptor-mediated transport system. Vitamin B 12 deficiency in humans most often results from malabsorption of vitamin B12 due either to lack of intrinsic factor or to loss or malfunction of the absorptive mechanism in the distal ileum. Nutritional deficiency is rare but may be seen in strict vegetarians after many years without meat, eggs, or dairy products. Once absorbed, vitamin B12 is transported to the various cells of the body bound to a family of specialized glycoproteins, transcobalamin I, II, and III. Excess vitamin B12 is stored in the liver.

Pharmacodynamics Two essential enzymatic reactions in humans require vitamin B 12 (Figure 33–2). In one, methylcobalamin serves as an intermediate in the transfer of a methyl group from N5 -methyltetrahydrofolate to homocysteine, forming methionine (Figure 33–2A; Figure 33–3, section 1). Without vitamin B 12 , conversion of the major dietary and storage folate—N5 -methyltetrahydrofolate—to tetrahydrofolate, the precursor of folate cofactors, cannot occur. As a result, vitamin B 12 deficiency leads to deficiency of folate cofactors necessary for several biochemical reactions involving the transfer of one-carbon groups. In particular, the depletion of tetrahydrofolate prevents synthesis of adequate supplies of the deoxythymidylate (dTMP) and purines required for DNA synthesis in rapidly dividing cells, as shown in Figure 33–3, section 2. The accumulation of folate as N5 -methyltetrahydrofolate and the associated depletion of tetrahydrofolate cofactors in vitamin B12 deficiency have been referred to as the “methylfolate trap.” This is the biochemical step whereby vitamin B12 and folic acid metabolism are linked, and it explains why the megaloblastic anemia of vitamin B12 deficiency can be partially corrected by ingestion of large amounts of folic acid. Folic acid can be reduced to dihydrofolate by the enzyme dihydrofolate reductase (Figure 33–3, section 3) and thereby serve as a source of the tetrahydrofolate required for synthesis of the purines and dTMP required for DNA synthesis.


FIGURE 33–2 Enzymatic reactions that use vitamin B12 . See text for details.


FIGURE 33–3 Enzymatic reactions that use folates. Section 1 shows the vitamin B12 -dependent reaction that allows most dietary folates to enter the tetrahydrofolate cofactor pool and becomes the “folate trap” in vitamin B12 deficiency. Section 2 shows the deoxythymidine monophosphate (dTMP) cycle. Section 3 shows the pathway by which folic acid enters the tetrahydrofolate cofactor pool. Double arrows indicate pathways with more than one intermediate step. dUMP, deoxyuridine monophosphate. Vitamin B12 deficiency causes the accumulation of homocysteine due to reduced formation of methylcobalamin, which is required for the conversion of homocysteine to methionine (Figure 33–3, section 1). The increase in serum homocysteine can be used to help establish a diagnosis of vitamin B12 deficiency (Table 33–2) . There is evidence from observational studies that elevated serum homocysteine increases the risk of atherosclerotic cardiovascular disease. However, randomized clinical trials have not shown a definitive reduction in


cardiovascular events (myocardial infarction, stroke) in patients receiving vitamin supplementation that lowers serum homocysteine. The other reaction that requires vitamin B12 is isomerization of methylmalonyl-CoA to succinyl-CoA by the enzyme methylmalonylCoA mutase (Figure 33–2B). In vitamin B12 deficiency, this conversion cannot take place and the substrate, methylmalonyl-CoA, as well as methylmalonic acid accumulate. The increase in serum and urine concentrations of methylmalonic acid can be used to support a diagnosis of vitamin B12 deficiency (Table 33–2). In the past, it was thought that abnormal accumulation of methylmalonyl-CoA causes the neurologic manifestations of vitamin B12 deficiency. However, newer evidence instead implicates the disruption of the methionine synthesis pathway as the cause of neurologic problems. Whatever the biochemical explanation for neurologic damage, the important point is that administration of folic acid in the setting of vitamin B12 deficiency will not prevent neurologic manifestations even though it will largely correct the anemia caused by the vitamin B12 deficiency.

Clinical Pharmacology Vitamin B 12 is used to treat or prevent deficiency. The most characteristic clinical manifestation of vitamin B 12 deficiency is megaloblastic, macrocytic anemia (Table 33–2), often with associated mild or moderate leukopenia or thrombocytopenia (or both), and a characteristic hypercellular bone marrow with an accumulation of megaloblastic erythroid and other precursor cells. The neurologic syndrome associated with vitamin B12 deficiency usually begins with paresthesias in peripheral nerves and weakness and progresses to spasticity, ataxia, and other central nervous system dysfunctions. Correction of vitamin B 12 deficiency arrests the progression of neurologic disease, but it may not fully reverse neurologic symptoms that have been present for several months. Although most patients with neurologic abnormalities caused by vitamin B12 deficiency have megaloblastic anemia when first seen, occasional patients have few if any hematologic abnormalities. Once a diagnosis of megaloblastic anemia is made, it must be determined whether vitamin B12 or folic acid deficiency is the cause. (Other causes of megaloblastic anemia are very rare.) This can usually be accomplished by measuring serum levels of the vitamins. The Schilling test, which measures absorption and urinary excretion of radioactively labeled vitamin B12 , can be used to further define the mechanism of vitamin B12 malabsorption when this is found to be the cause of the megaloblastic anemia. The most common causes of vitamin B12 deficiency are pernicious anemia, partial or total gastrectomy, and conditions that affect the distal ileum, such as malabsorption syndromes, inflammatory bowel disease, or small bowel resection. Pernicious anemia results from defective secretion of intrinsic factor by the gastric mucosal cells. Patients with pernicious anemia have gastric atrophy and fail to secrete intrinsic factor (as well as hydrochloric acid). These patients frequently have autoantibodies to intrinsic factor. The Schilling test shows diminished absorption of radioactively labeled vitamin B 12 , which is corrected when intrinsic factor is administered with radioactive B12 , since the vitamin can then be normally absorbed. Vitamin B 12 deficiency also occurs when the region of the distal ileum that absorbs the vitamin B12 -intrinsic factor complex is damaged, as when the ileum is involved with inflammatory bowel disease or when the ileum is surgically resected. In these situations, radioactively labeled vitamin B12 is not absorbed in the Schilling test, even when intrinsic factor is added. Rare cases of vitamin B12 deficiency in children have been found to be secondary to congenital deficiency of intrinsic factor or to defects of the receptor sites for vitamin B12 -intrinsic factor complex located in the distal ileum. Because it is associated with use of radioactive isotopes, the Schilling test is unavailable in many centers. Alternatively one can test for intrinsic factor antibodies, and for elevated homocysteine and methylmalonic acid levels (Figure 33–2) to make a diagnosis of pernicious anemia with high sensitivity and specificity. Almost all cases of vitamin B12 deficiency are caused by malabsorption of the vitamin; therefore, parenteral injections of vitamin B12 are required for therapy. For patients with potentially reversible diseases, the underlying disease should be treated after initial treatment with parenteral vitamin B12 . Most patients, however, do not have curable deficiency syndromes and require lifelong treatment with vitamin B12 . Vitamin B12 for parenteral injection is available as cyanocobalamin or hydroxocobalamin. Hydroxocobalamin is preferred because it is more highly protein-bound and therefore remains longer in the circulation. Initial therapy should consist of 100–1000 mcg of vitamin B12 intramuscularly daily or every other day for 1–2 weeks to replenish body stores. Maintenance therapy consists of 100–1000 mcg intramuscularly once a month for life. If neurologic abnormalities are present, maintenance therapy injections should be given every 1–2 weeks for 6 months before switching to monthly injections. Oral vitamin B12 -intrinsic factor mixtures and liver extracts should not be used to treat vitamin B12 deficiency; however, oral doses of 1000 mcg of vitamin B 12 daily are usually sufficient to treat patients with pernicious anemia who refuse or cannot tolerate the injections. After pernicious anemia is in remission following parenteral vitamin B 12 therapy, the vitamin can be administered intranasally as a spray or gel.

FOLIC ACID Reduced forms of folic acid are required for essential biochemical reactions that provide precursors for the synthesis of amino acids,


purines, and DNA. Folate deficiency is relatively common, even though the deficiency is easily corrected by administration of folic acid. The consequences of folate deficiency go beyond the problem of anemia because folate deficiency is implicated as a cause of congenital malformations in newborns and may play a role in vascular disease (see Box: Folic Acid Supplementation: A Public Health Dilemma).

Chemistry Folic acid (pteroylglutamic acid) is composed of a heterocycle (pteridine), p-aminobenzoic acid, and glutamic acid (Figure 33–4). Various numbers of glutamic acid moieties are attached to the pteroyl portion of the molecule, resulting in monoglutamates, triglutamates, or polyglutamates. Folic acid undergoes reduction, catalyzed by the enzyme dihydrofolate reductase (“folate reductase”), to give dihydrofolic acid (Figure 33–3, section 3). Tetrahydrofolate is subsequently transformed to folate cofactors possessing one-carbon units attached to the 5-nitrogen, to the 10-nitrogen, or to both positions (Figure 33–3). Folate cofactors are interconvertible by various enzymatic reactions and serve the important biochemical function of donating one-carbon units at various levels of oxidation. In most of these, tetrahydrofolate is regenerated and becomes available for reutilization.

FIGURE 33–4 The structure of folic acid. (Reproduced, with permission, from Murray RK et al: Harper’s Biochemistry, 24th ed. McGraw-Hill, 1996. Copyright © The McGraw-Hill Companies, Inc.)

Folic Acid Supplementation: A Public Health Dilemma Starting in January 1998, all products made from enriched grains in the United States and Canada were required to be supplemented with folic acid. These rulings were issued to reduce the incidence of congenital neural tube defects (NTDs). Epidemiologic studies show a strong correlation between maternal folic acid deficiency and the incidence of NTDs such as spina bifida and anencephaly. The requirement for folic acid supplementation is a public health measure aimed at the significant number of women who do not receive prenatal care and are not aware of the importance of adequate folic acid ingestion for preventing birth defects in their infants. Observational studies from countries that supplement grains with folic acid have found that supplementation is associated with a significant (20–25%) reduction in NTD rates. Observational studies also suggest that rates of other types of congenital anomalies (heart and orofacial) have fallen since supplementation began. There may be an added benefit for adults. N5 -Methyl-tetrahydrofolate is required for the conversion of homocysteine to methionine (Figure 33–2; Figure 33–3, reaction 1). Impaired synthesis of N5 -methyltetrahydrofolate results in elevated serum concentrations of homocysteine. Data from several sources suggest a positive correlation between elevated serum homocysteine and occlusive vascular diseases such as ischemic heart disease and stroke. Clinical data suggest that the folate supplementation program has improved the folate status and reduced the prevalence of hyperhomocysteinemia in a population of middle-aged and older adults who did not use vitamin supplements. There is also evidence that adequate folic acid protects against several cancers, including colorectal, breast, and cervical cancer. Although the potential benefits of supplemental folic acid during pregnancy are compelling, the decision to require folic acid in grains was controversial. As described in the text, ingestion of folic acid can partially or totally correct the anemia caused by vitamin B12 deficiency. However, folic acid supplementation does not prevent the potentially irreversible neurologic damage caused by


vitamin B12 deficiency. People with pernicious anemia and other forms of vitamin B 12 deficiency are usually identified because of signs and symptoms of anemia, which typically occur before neurologic symptoms. Some opponents of folic acid supplementation were concerned that increased folic acid intake in the general population would mask vitamin B12 deficiency and increase the prevalence of neurologic disease in the elderly population. To put this in perspective, approximately 4000 pregnancies, including 2500 live births, in the United States each year are affected by NTDs. In contrast, it is estimated that over 10% of the elderly population in the United States, or several million people, are at risk for the neuropsychiatric complications of vitamin B12 deficiency. In acknowledgment of this controversy, the FDA kept its requirements for folic acid supplementation at a somewhat low level. There is also concern based on observational and prospective clinical trials that high folic acid levels can increase the risk of some diseases, such as colorectal cancer, for which folic acid may exhibit a bell-shaped curve. Further research is needed to more accurately define the optimal level of folic acid fortification in food and recommendations for folic acid supplementation in different populations and age groups.

Pharmacokinetics The average American diet contains 500–700 mcg of folates daily, 50–200 mcg of which is usually absorbed, depending on metabolic requirements. Pregnant women may absorb as much as 300–400 mcg of folic acid daily. Various forms of folic acid are present in a wide variety of plant and animal tissues; the richest sources are yeast, liver, kidney, and green vegetables. Normally, 5–20 mg of folates is stored in the liver and other tissues. Folates are excreted in the urine and stool and are also destroyed by catabolism, so serum levels fall within a few days when intake is diminished. Because body stores of folates are relatively low and daily requirements high, folic acid deficiency and megaloblastic anemia can develop within 1–6 months after the intake of folic acid stops, depending on the patient’s nutritional status and the rate of folate utilization. Unaltered folic acid is readily and completely absorbed in the proximal jejunum. Dietary folates, however, consist primarily of polyglutamate forms of N5 -methyltetrahydrofolate. Before absorption, all but one of the glutamyl residues of the polyglutamates must be hydrolyzed by the enzyme α-1-glutamyl transferase (“conjugase”) within the brush border of the intestinal mucosa. The monoglutamate N5 -methyltetrahydrofolate is subsequently transported into the bloodstream by both active and passive transport and is then widely distributed throughout the body. Inside cells, N5 -methyltetrahydro-folate is converted to tetrahydrofolate by the demethylation reaction that requires vitamin B12 (Figure 33–3, section 1).

Pharmacodynamics Tetrahydrofolate cofactors participate in one-carbon transfer reactions. As described earlier in the discussion of vitamin B 12 , one of these essential reactions produces the dTMP needed for DNA synthesis. In this reaction, the enzyme thymidylate synthase catalyzes the transfer of the one-carbon unit of N5 , N10 -methylenetetrahydrofolate to deoxyuridine monophosphate (dUMP) to form dTMP (Figure 33–3, section 2). Unlike all the other enzymatic reactions that use folate cofactors, in this reaction the cofactor is oxidized to dihydrofolate, and for each mole of dTMP produced, 1 mole of tetrahydrofolate is consumed. In rapidly proliferating tissues, considerable amounts of tetrahydrofolate are consumed in this reaction, and continued DNA synthesis requires continued regeneration of tetrahydrofolate by reduction of dihydrofolate, catalyzed by the enzyme dihydrofolate reductase. The tetrahydrofolate thus produced can then reform the cofactor N5 , N10 -methylenetetrahydrofolate by the action of serine transhydroxymethylase and thus allow for the continued synthesis of dTMP. The combined catalytic activities of dTMP synthase, dihydrofolate reductase, and serine transhydroxymethylase are referred to as the dTMP synthesis cycle. Enzymes in the dTMP cycle are the targets of two anti-cancer drugs; methotrexate inhibits dihydrofolate reductase, and a metabolite of 5-fluorouracil inhibits thymidylate synthase (see Chapter 54). Cofactors of tetrahydrofolate participate in several other essential reactions. N5 -Methylenetetrahydrofolate is required for the vitamin B12 -dependent reaction that generates methionine from homocysteine (Figure 33–2A; Figure 33–3, section 1). In addition, tetrahydrofolate cofactors donate one-carbon units during the de novo synthesis of essential purines. In these reactions, tetrahydrofolate is regenerated and can reenter the tetrahydrofolate cofactor pool.

Clinical Pharmacology Folate deficiency results in a megaloblastic anemia that is microscopically indistinguishable from the anemia caused by vitamin B12 deficiency (see above). However, folate deficiency does not cause the characteristic neurologic syndrome seen in vitamin B 12 deficiency. In patients with megaloblastic anemia, folate status is assessed with assays for serum folate or for red blood cell folate. Red blood cell folate levels are often of greater diagnostic value than serum levels, because serum folate levels tend to be labile and do not necessarily reflect tissue levels. Folic acid deficiency is often caused by inadequate dietary intake of folates. Patients with alcohol dependence and patients with liver disease can develop folic acid deficiency because of poor diet and diminished hepatic storage of folates. Pregnant women and patients


with hemolytic anemia have increased folate requirements and may become folic acid-deficient, especially if their diets are marginal. Evidence implicates maternal folic acid deficiency in the occurrence of fetal neural tube defects. (See Box: Folic Acid Supplementation: A Public Health Dilemma.) Patients with malabsorption syndromes also frequently develop folic acid deficiency. Patients who require renal dialysis are at risk of folic acid deficiency because folates are removed from the plasma during the dialysis procedure. Folic acid deficiency can be caused by drugs. Methotrexate and, to a lesser extent, trimethoprim and pyrimethamine, inhibit dihydrofolate reductase and may result in a deficiency of folate cofactors and ultimately in megaloblastic anemia. Long-term therapy with phenytoin can also cause folate deficiency, but only rarely causes megaloblastic anemia. Parenteral administration of folic acid is rarely necessary, since oral folic acid is well absorbed even in patients with malabsorption syndromes. A dose of 1 mg folic acid orally daily is sufficient to reverse megaloblastic anemia, restore normal serum folate levels, and replenish body stores of folates in almost all patients. Therapy should be continued until the underlying cause of the deficiency is removed or corrected. Therapy may be required indefinitely for patients with malabsorption or dietary inadequacy. Folic acid supplementation to prevent folic acid deficiency should be considered in high-risk patients, including pregnant women, patients with alcohol dependence, hemolytic anemia, liver disease, or certain skin diseases, and patients on renal dialysis.

HEMATOPOIETIC GROWTH FACTORS The hematopoietic growth factors are glycoprotein hormones that regulate the proliferation and differentiation of hematopoietic progenitor cells in the bone marrow. The first growth factors to be identified were called colony-stimulating factors because they could stimulate the growth of colonies of various bone marrow progenitor cells in vitro. Many of these growth factors have been purified and cloned, and their effects on hematopoiesis have been extensively studied. Quantities of these growth factors sufficient for clinical use are produced by recombinant DNA technology. Of the known hematopoietic growth factors, erythropoietin (epoetin alfa and epoetin beta), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-11 (IL-11) and thrombopoietin receptor agonists (romiplostim and eltrombopag) are currently in clinical use. The hematopoietic growth factors and drugs that mimic their action have complex effects on the function of a wide variety of cell types, including nonhematologic cells. Their usefulness in other areas of medicine, particularly as potential anti-cancer and antiinflammatory drugs, is being investigated.

ERYTHROPOIETIN Chemistry & Pharmacokinetics Erythropoietin, a 34–39 kDa glycoprotein, was the first human hematopoietic growth factor to be isolated. It was originally purified from the urine of patients with severe anemia. Recombinant human erythropoietin (rHuEPO, epoetin alfa) is produced in a mammalian cell expression system. After intravenous administration, erythropoietin has a serum half-life of 4–13 hours in patients with chronic renal failure. It is not cleared by dialysis. It is measured in international units (IU). Darbepoetin alfa is a modified form of erythropoietin that is more heavily glycosylated as a result of changes in amino acids. Darbepoetin alfa has a twofold to threefold longer half-life than epoetin alfa. Methoxy polyethylene glycol-epoetin beta is an isoform of erythropoietin covalently attached to a long polyethylene glycol polymer. This long-lived recombinant product is administered as a single intravenous or subcutaneous dose at 2-week or monthly intervals, whereas epoetin alfa is generally administered three times a week and darbepoetin is administered weekly.

Pharmacodynamics Erythropoietin stimulates erythroid proliferation and differentiation by interacting with erythropoietin receptors on red cell progenitors. The erythropoietin receptor is a member of the JAK/STAT superfamily of cytokine receptors that use protein phosphorylation and transcription factor activation to regulate cellular function (see Chapter 2). Erythropoietin also induces release of reticulocytes from the bone marrow. Endogenous erythropoietin is primarily produced in the kidney. In response to tissue hypoxia, more erythropoietin is produced through an increased rate of transcription of the erythropoietin gene. This results in correction of the anemia, provided that the bone marrow response is not impaired by red cell nutritional deficiency (especially iron deficiency), primary bone marrow disorders (see below), or bone marrow suppression from drugs or chronic diseases. Normally, an inverse relationship exists between the hematocrit or hemoglobin level and the serum erythropoietin level. Nonanemic individuals have serum erythropoietin levels of less than 20 IU/L. As the hematocrit and hemoglobin levels fall and anemia becomes more severe, the serum erythropoietin level rises exponentially. Patients with moderately severe anemia usually have erythropoietin levels in the 100–500 IU/L range, and patients with severe anemia may have levels of thousands of IU/L. The most important exception to this inverse relationship is in the anemia of chronic renal failure. In patients with renal disease, erythropoietin levels are usually low because the kidneys cannot produce the growth factor. These are the patients most likely to respond to treatment with exogenous erythropoietin.


In most primary bone marrow disorders (aplastic anemia, leukemias, myeloproliferative and myelodysplastic disorders, etc) and most nutritional and secondary anemias, endogenous erythropoietin levels are high, so there is less likelihood of a response to exogenous erythropoietin (but see below).

Clinical Pharmacology The availability of erythropoiesis-stimulating agents (ESAs) has had a significant positive impact for patients with several types of anemia (Table 33–4). The ESAs consistently improve the hematocrit and hemoglobin level, often eliminate the need for transfusions, and reliably improve quality of life indices. The ESAs are used routinely in patients with anemia secondary to chronic kidney disease. In patients treated with an ESA, an increase in reticulocyte count is usually observed in about 10 days and an increase in hematocrit and hemoglobin levels in 2–6 weeks. Dosages of ESAs are adjusted to maintain a target hemoglobin up to, but not exceeding, 10–12 g/dL. To support the increased erythropoiesis, nearly all patients with chronic kidney disease require oral or parenteral iron supplementation. Folate supplementation may also be necessary in some patients. TABLE 33–4 Clinical uses of hematopoietic growth factors and agents that mimic their actions.

In selected patients, erythropoietin is also used to reduce the need for red blood cell transfusion in patients undergoing myelosuppressive cancer chemotherapy who have a hemoglobin level of less than 10 g/dL, and for selected patients with low-risk


myelodysplastic syndromes and anemia requiring red blood cell transfusion. Patients who have disproportionately low serum erythropoietin levels for their degree of anemia are most likely to respond to treatment. Patients with endogenous erythropoietin levels of less than 100 IU/L have the best chance of response, although patients with erythropoietin levels between 100 and 500 IU/L respond occasionally. Methoxy polyethylene glycol-epoetin beta should not be used for treatment of anemia caused by cancer chemotherapy because a clinical trial found significantly more deaths among patients receiving this form of erythropoietin. Erythropoietin is one of the drugs commonly used illegally by endurance athletes to enhance performance. Other methods such as autologous transfusion of red cells or use of androgens have also been used to increase hemoglobin. “Blood doping” constitutes a serious health risk to athletes and as a form of cheating is universally banned and routinely tested for in athletic events.

Toxicity The most common adverse effects of erythropoietin are hypertension and thrombotic complications. ESAs increase the risk of serious cardiovascular events, thromboembolic events, stroke, and mortality in clinical studies when given to support hemoglobin levels greater than 11 g/dL In addition, a meta-analysis of 51 placebo-controlled trials of ESAs in cancer patients reported an increased rate of allcause mortality and venous thrombosis in those receiving an ESA. Based on the accumulated evidence, it is recommended that the hemoglobin level not exceed 11 g/dL in patients with chronic kidney disease receiving an ESA, and that ESAs be used conservatively in cancer patients (eg, when hemoglobin levels are < 10 g/dL) and with the lowest dose needed to avoid transfusion. It is further recommended that ESAs not be used when a cancer therapy is being given with curative intent. Allergic reactions to ESAs have been infrequent. There have been a small number of cases of pure red cell aplasia (PRCA) accompanied by neutralizing antibodies to erythropoietin. PRCA was most commonly seen in dialysis patients treated subcutaneously for a long period with a particular form of epoetin alfa (Eprex with a polysorbate 80 stabilizer rather than human serum albumin) that is not available in the United States. After regulatory agencies required that Eprex be administered intravenously rather than subcutaneously, the rate of ESA-associated PRCA diminished. However, rare cases have still been seen with all ESAs administered subcutaneously for long periods to patients with chronic kidney disease.

MYELOID GROWTH FACTORS Chemistry & Pharmacokinetics G-CSF and GM-CSF, the two myeloid growth factors currently available for clinical use, were originally purified from cultured human cell lines (Table 33–4). Recombinant human G-CSF (rHuG-CSF; filgrastim) is produced in a bacterial expression system. It is a nonglycosylated peptide of 175 amino acids, with a molecular weight of 18 kDa. Recombinant human GM-CSF (rHuGM-CSF; sargramostim) is produced in a yeast expression system. It is a partially glycosylated peptide of 127 amino acids, comprising three molecular species with molecular weights of 15,500, 15,800, and 19,500. These preparations have serum half-lives of 2–7 hours after intravenous or subcutaneous administration. Pegfilgrastim, a covalent conjugation product of filgrastim and a form of polyethylene glycol, has a much longer serum half-life than recombinant G-CSF, and it can be injected once per myelosuppressive chemotherapy cycle instead of daily for several days. Lenograstim, used widely in Europe, is a glycosylated form of recombinant G-CSF.

Pharmacodynamics The myeloid growth factors stimulate proliferation and differentiation by interacting with specific receptors found on myeloid progenitor cells. Like the erythropoietin receptor, these receptors are members of the JAK/STAT superfamily (see Chapter 2). G-CSF stimulates proliferation and differentiation of progenitors already committed to the neutrophil lineage. It also activates the phagocytic activity of mature neutrophils and prolongs their survival in the circulation. G-CSF also has a remarkable ability to mobilize hematopoietic stem cells, ie, to increase their concentration in peripheral blood. This biologic effect underlies a major advance in transplantation—the use of peripheral blood stem cells (PBSCs) rather than bone marrow stem cells for autologous and allogeneic hematopoietic stem cell transplantation (see below). GM-CSF has broader biologic actions than G-CSF. It is a multipotential hematopoietic growth factor that stimulates proliferation and differentiation of early and late granulocytic progenitor cells as well as erythroid and megakaryocyte progenitors. Like G-CSF, GM-CSF also stimulates the function of mature neutrophils. GM-CSF acts together with interleukin-2 to stimulate T-cell proliferation and appears to be a locally active factor at the site of inflammation. GM-CSF mobilizes peripheral blood stem cells, but it is significantly less efficacious and more toxic than G-CSF in this regard.

Clinical Pharmacology A. Cancer Chemotherapy-Induced Neutropenia


Neutropenia is a common adverse effect of the cytotoxic drugs used to treat cancer and increases the risk of serious infection in patients receiving chemotherapy. Unlike the treatment of anemia and thrombocytopenia, transfusion of neutropenic patients with granulocytes collected from donors is performed rarely and with limited success. The introduction of G-CSF in 1991 represented a milestone in the treatment of chemotherapy-induced neutropenia. This growth factor dramatically accelerates the rate of neutrophil recovery after doseintensive myelosuppressive chemotherapy (Figure 33–5). It reduces the duration of neutropenia and usually raises the nadir count, the lowest neutrophil count seen following a cycle of chemotherapy.

FIGURE 33–5 Effects of granulocyte colony-stimulating factor (G-CSF; red line) or placebo (green line) on absolute neutrophil count (ANC) after cytotoxic chemotherapy for lung cancer. Doses of chemotherapeutic drugs were administered on days 1 and 3. G-CSF or placebo injections were started on day 4 and continued daily through day 12 or 16. The first peak in ANC reflects the recruitment of mature cells by G-CSF. The second peak reflects a marked increase in new neutrophil production by the bone marrow under stimulation by G-CSF. (Normal ANC is 2.2–8.6 × 109 /L.) (Reproduced, with permission, from Crawford J et al: Reduction by granulocyte colonystimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer. N Engl J Med 1991;325:164. Copyright © 1991 Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society.) The ability of G-CSF to increase neutrophil counts after myelosuppressive chemotherapy is nearly universal, but its impact on clinical outcomes is more variable. Many, but not all, clinical trials and meta-analyses have shown that G-CSF reduces episodes of febrile neutropenia, requirements for broad-spectrum antibiotics, infections, and days of hospitalization. Clinical trials have not shown improved survival in cancer patients treated with G-CSF. Clinical guidelines for the use of G-CSF after cytotoxic chemotherapy recommend reserving G-CSF for patients at high risk for febrile neutropenia based on age, medical history, and disease characteristics; patients receiving dose-intensive chemotherapy regimens that carry a greater than 20% risk of causing febrile neutropenia; patients with a prior episode of febrile neutropenia after cytotoxic chemotherapy; patients at high risk for febrile neutropenia; and patients who are unlikely to survive an episode of febrile neutropenia. Pegfilgrastim is an alternative to G-CSF for prevention of chemotherapy-induced febrile neutropenia. Pegfilgrastim can be administered once per chemotherapy cycle, and it may shorten the period of severe neutropenia slightly more than G-CSF. Like G-CSF and pegfilgrastim, GM-CSF also reduces the duration of neutropenia after cytotoxic chemotherapy. It has been more difficult to show that GM-CSF reduces the incidence of febrile neutropenia, probably because GM-CSF itself can induce fever. In the treatment of chemotherapy-induced neutropenia, G-CSF, 5 mcg/kg/d, or GM-CSF, 250 mcg/m 2 /d, is usually started within 24–72 hours after completing chemotherapy and is continued until the absolute neutrophil count is greater than 10,000 cells/μL. Pegfilgrastim is given as a single dose of 6 mg. The utility and safety of the myeloid growth factors in the postchemotherapy supportive care of patients with acute myeloid leukemia (AML) have been the subject of a number of clinical trials. Because leukemic cells arise from progenitors whose proliferation and differentiation are normally regulated by hematopoietic growth factors, including GM-CSF and G-CSF, there was concern that myeloid growth factors could stimulate leukemic cell growth and increase the rate of relapse. The results of randomized clinical trials suggest that both G-CSF and GM-CSF are safe following induction and consolidation treatment of myeloid and lymphoblastic leukemia. There has been no evidence that these growth factors reduce the rate of remission or increase relapse rate. On the contrary, the growth factors accelerate neutrophil recovery and reduce infection rates and days of hospitalization. Both G-CSF and GM-CSF have FDA approval for treatment of patients with AML. B. Other Applications G-CSF and GM-CSF have also proved to be effective in treating the neutropenia associated with congenital neutropenia, cyclic neutropenia, myelodysplasia, and aplastic anemia. Many patients with these disorders respond with a prompt and sometimes dramatic increase in neutrophil count. In some cases, this results in a decrease in the frequency of infections. Because neither G-CSF


nor GM-CSF stimulates the formation of erythrocytes and platelets, they are sometimes combined with other growth factors for treatment of pancytopenia. The myeloid growth factors play an important role in autologous stem cell transplantation for patients undergoing high-dose chemotherapy. High-dose chemotherapy with autologous stem cell support is increasingly used to treat patients with tumors that are resistant to standard doses of chemotherapeutic drugs. The high-dose regimens produce extreme myelosuppression; the myelosuppression is then counteracted by reinfusion of the patient’s own hematopoietic stem cells (which are collected prior to chemotherapy). The administration of G-CSF or GM-CSF early after autologous stem cell transplantation reduces the time to engraftment and to recovery from neutropenia in patients receiving stem cells obtained either from bone marrow or from peripheral blood. These effects are seen in patients being treated for lymphoma or for solid tumors. G-CSF and GM-CSF are also used to support patients who have received allogeneic bone marrow transplantation for treatment of hematologic malignancies or bone marrow failure states. In this setting, the growth factors speed the recovery from neutropenia without increasing the incidence of acute graft-versus-host disease. Perhaps the most important role of the myeloid growth factors in transplantation is for mobilization of PBSCs. Stem cells collected from peripheral blood have nearly replaced bone marrow as the hematopoietic preparation used for autologous and allogeneic transplantation. The cells can be collected in an outpatient setting with a procedure that avoids much of the risk and discomfort of bone marrow collection, including the need for general anesthesia. In addition, there is evidence that PBSC transplantation results in more rapid engraftment of all hematopoietic cell lineages and in reduced rates of graft failure or delayed platelet recovery. G-CSF is the cytokine most commonly used for PBSC mobilization because of its increased efficacy and reduced toxicity compared with GM-CSF. To mobilize stem cells for autologous transplantation, donors are given 5–10 mcg/kg/d subcutaneously for 4 days. On the fifth day, they undergo leukapheresis. The success of PBSC transplantation depends on transfusion of adequate numbers of stem cells. CD34, an antigen present on early progenitor cells and absent from later, committed, cells, is used as a marker for the requisite stem cells. The goal is to infuse at least 5 × 106 CD34 cells/kg; this number of CD34 cells usually results in prompt and durable engraftment of all cell lineages. It may take several separate leukaphereses to collect enough CD34 cells, especially from older patients and patients who have been exposed to radiation therapy or chemotherapy. For patients with multiple myeloma or non-Hodgkin’s lymphoma who respond suboptimally to G-CSF alone, the novel hematopoietic stem cell mobilizer plerixafor can be added to G-CSF. Plerixafor is a bicyclam molecule originally developed as an anti-HIV drug because of its ability to inhibit the CXC chemokine receptor 4 (CXCR4), a co-receptor for HIV entry into CD4+ T lymphocytes (see Chapter 49). Early clinical trials of plerixafor revealed a remarkable ability to increase CD34 cells in peripheral blood. Plerixafor mobilizes CD34 cells by preventing chemokine stromal cell-derived factor-1α (SDF-1α) from binding to CXCR4 and directing the CD34 cells to “home” to the bone marrow. Plerixafor is administered by subcutaneous injection after 4 days of G-CSF treatment and 11 hours prior to leukapheresis; it can be used with G-CSF for up to 4 continuous days. Plerixafor is eliminated primarily by the renal route and must be dose-adjusted for patients with renal impairment. The drug is well-tolerated; the most common adverse effects associated with its use are injection site reactions, gastrointestinal disturbances, dizziness, fatigue, and headache.

Toxicity Although the three growth factors have similar effects on neutrophil counts, G-CSF and pegfilgrastim are used more frequently than GMCSF because they are better tolerated. G-CSF and pegfilgrastim can cause bone pain, which clears when the drugs are discontinued. GM-CSF can cause more severe side effects, particularly at higher doses. These include fever, malaise, arthralgias, myalgias, and a capillary leak syndrome characterized by peripheral edema and pleural or pericardial effusions. Allergic reactions may occur but are infrequent. Splenic rupture is a rare but serious complication of the use of G-CSF for PBSC.

MEGAKARYOCYTE GROWTH FACTORS Patients with thrombocytopenia have a high risk of hemorrhage. Although platelet transfusion is commonly used to treat thrombocytopenia, this procedure can cause adverse reactions in the recipient; furthermore, a significant number of patients fail to exhibit the expected increase in platelet count. Thrombopoietin (TPO) and IL-11 both appear to be key endogenous regulators of platelet production. A recombinant form of IL-11 was the first agent to gain FDA approval for treatment of thrombocytopenia. Recombinant human thrombopoietin and a pegylated form of a shortened human thrombopoietin protein underwent extensive clinical investigation in the 1990s. However, further development was abandoned after autoantibodies to the native thrombopoietin formed in healthy human subjects and caused thrombocytopenia. Efforts shifted to investigation of novel, nonimmunogenic agonists of the thrombopoietin receptor, which is known as Mpl. Two thrombopoietin agonists (romiplostim and eltrombopag) are approved for treatment of thrombocytopenia.

Chemistry & Pharmacokinetics Interleukin-11 is a 65–85 kDa protein produced by fibroblasts and stromal cells in the bone marrow. Oprelvekin, the recombinant


form of IL-11 approved for clinical use (Table 33–4), is produced by expression in Escherichia coli. The half-life of IL-11 is 7–8 hours when the drug is injected subcutaneously. Romiplostim is a peptide covalently linked to antibody fragments that serve to extend the peptide’s half-life. The Mpl-binding peptide has no sequence homology with human thrombopoietin and there is no evidence in animal or human studies that the Mpl-binding peptide or romiplostim induces antibodies to thrombopoietin. After subcutaneous administration, romiplostim is eliminated by the reticuloendothelial system with an average half-life of 3–4 days. Its half-life is inversely related to the serum platelet count; it has a longer half-life in patients with thrombocytopenia and a shorter half-life in patients whose platelet counts have recovered to normal levels. Romiplostim is approved for therapy of patients with chronic immune thrombocytopenia who have had an inadequate response to other therapies. Eltrombopag is an orally active small nonpeptide thrombopoietin agonist molecule approved for therapy of patients with chronic immune thrombocytopenia who have had an inadequate response to other therapies, and for treatment of thrombocytopenia in patients with hepatitis C to allow initiation of interferon therapy. Following oral administration, peak eltrombopag levels are observed in 2–6 hours and half-life is 26–35 hours. Eltrombopag is primarily excreted in the feces.

Pharmacodynamics Interleukin-11 acts through a specific cell surface cytokine receptor to stimulate the growth of multiple lymphoid and myeloid cells. It acts synergistically with other growth factors to stimulate the growth of primitive megakaryocytic progenitors and, most importantly, increases the number of peripheral platelets and neutrophils. Romiplostim has high affinity for the human Mpl receptor. Eltrombopag interacts with the transmembrane domain of the Mpl receptor. Both drugs induce signaling through the Mpl receptor pathway and cause a dose-dependent increase in platelet count. Romiplostim is administered once weekly by subcutaneous injection. Eltrombopag is an oral drug. For both drugs, peak platelet count responses are observed in approximately 2 weeks.

Clinical Pharmacology Interleukin-11 is approved for the secondary prevention of thrombocytopenia in patients receiving cytotoxic chemotherapy for treatment of nonmyeloid cancers. Clinical trials show that it reduces the number of platelet transfusions required by patients who experience severe thrombocytopenia after a previous cycle of chemotherapy. Although IL-11 has broad stimulatory effects on hematopoietic cell lineages in vitro, it does not appear to have significant effects on the leukopenia caused by myelosuppressive chemotherapy. Interleukin-11 is given by subcutaneous injection at a dose of 50 mcg/kg/d. It is started 6–24 hours after completion of chemotherapy and continued for 14–21 days or until the platelet count passes the nadir and rises to more than 50,000 cells/μL. In patients with chronic immune thrombocytopenia who failed to respond adequately to previous treatment with steroids, immunoglobulins, or splenectomy, romiplostim and eltrombopag significantly increase platelet count in most patients. Both drugs are used at the minimal dose required to maintain platelet counts of greater than 50,000 cells/μL.

Toxicity The most common adverse effects of IL-11 are fatigue, headache, dizziness, and cardiovascular effects. The cardiovascular effects include anemia (due to hemodilution), dyspnea (due to fluid accumulation in the lungs), and transient atrial arrhythmias. Hypokalemia has also been seen in some patients. All of these adverse effects appear to be reversible. Eltrombopag is potentially hepatotoxic and liver function must be monitored, particularly when used in patients with hepatitis C. Portal vein thrombosis has also been reported with eltrombopag and romiplostim in the setting of chronic liver disease. In patients with myelodysplastic syndromes, romiplostim increases the blast count and risk of progression to acute myeloid leukemia. Marrow fibrosis has been observed with thrombopoietin agonists but is generally reversible when the drug is discontinued. Rebound thrombocytopenia has been observed following discontinuation of TPO agonists.

SUMMARY Agents Used in Anemias and Hematopoietic Growth Factors




PREPARATIONS AVAILABLE

REFERENCES Aapro MS et al, European Organisation for Research and T reatment of Cancer: 2010 update of EORT C guidelines for the use of granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-induced febrile neutropenia in adult patients with lymphoproliferative disorders and solid tumours. Eur J Cancer 2011;47:8. Albaramki J et al: Parenteral versus oral iron therapy for adults and children with chronic kidney disease. Cochrane Database Syst Rev 2012;(1):CD007857. Auerbach M, Al T alib K: Low-molecular weight iron dextran and iron sucrose have similar comparative safety profiles in chronic kidney disease. Kidney Int 2008;73:528. Barzi A, Sekeres MA: Myelodysplastic syndromes: A practical approach to diagnosis and treatment. Cleve Clin J Med 2010;77:37. Brittenham GM: Iron-chelating therapy for transfusional iron overload. N Engl J Med 2011;364:146. Clark SF: Iron deficiency anemia: diagnosis and management. Curr Opin Gastroenterol 2009;25:122. Darshan D, Fraer DM, Anderson GJ: Molecular basis of iron-loading disorders. Expert Rev Mol Med 2010;12:e36. Gertz MA: Current status of stem cell mobilization. Br J Haematol 2010;150:647. Kessans MR, Gatesman ML, Kockler DR: Plerixafor: A peripheral blood stem cell mobilizer. Pharmacotherapy 2010;30:485. McKoy JM et al: Epoetin-associated pure red cell aplasia: Past, present, and future considerations. T ransfusion 2008;48:1754. Rees DC, Williams T N, Gladwin MT : Sickle-cell disease. Lancet 2010;376:2018. Rizzo JD et al: American Society of Clinical Oncology/American Society of Hematology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. J Clin Oncol 2010;28:4996. Sauer J, Mason JB, Choi SW: T oo much folate: A risk factor for cancer and cardiovascular disease? Curr Opin Clin Nutr Metab Care 2009;12:30. Solomon LR: Disorders of cobalamin (vitamin B12) metabolism: Emerging concepts in pathophysiology, diagnosis and treatment. Blood Rev 2007;21:113. Stasi R et al: T hrombopoietic agents. Blood Rev 2010;24:179.


Wolff T et al: Folic acid supplementation for the prevention of neural tube defects: An update of the evidence for the U.S. Preventive Services T ask Force. Ann Intern Med 2009;150:632.

CASE STUDY ANSWER This patient’s megaloblastic anemia appears to be due to vitamin B 12 (cobalamin) deficiency secondary to impaired production of intrinsic factor, resulting in insufficient absorption of vitamin B 12 from the gastrointestinal tract. It is important to measure serum concentrations of both folic acid and cobalamin because megaloblastic anemia can result from deficiency of either nutrient. It is especially important to diagnose vitamin B12 deficiency because this deficiency, if untreated, can lead to irreversible neurologic damage. Folate supplementation, which can compensate for vitamin B12 -derived anemia, does not prevent B12 -deficiency neurologic damage. To correct this patient’s vitamin B 12 deficiency, she would probably be treated parenterally with cobalamin because of her impaired oral absorption of vitamin B12 . Several weeks of daily administration would be followed with weekly doses until her hematocrit returned to normal. Monthly doses would then be given to maintain her body stores of vitamin B12 .


_______________ * T he author acknowledges contributions of the previous author of this chapter, Susan B. Masters, PhD.


CHAPTER

34 Drugs Used in Disorders of Coagulation James L. Zehnder, MD

CASE STUDY A 25-year-old woman presents to the emergency department complaining of acute onset of shortness of breath and pleuritic pain. She had been in her usual state of health until 2 days prior when she noted that her left leg was swollen and red. Her only medication was oral contraceptives. Family history was significant for a history of “blood clots” in multiple members of the maternal side of her family. Physical examination demonstrates an anxious woman with stable vital signs. The left lower extremity demonstrates erythema and edema and is tender to touch. Ultrasound reveals a deep vein thrombosis in the left lower extremity; chest computed tomography scan confirms the presence of pulmonary emboli. Laboratory blood tests indicate elevated D-dimer levels. What therapy is indicated acutely? What are the long-term therapy options? How long should she be treated? Should this individual use oral contraceptives?

Hemostasis refers to the finely regulated dynamic process of maintaining fluidity of the blood, repairing vascular injury, and limiting blood loss while avoiding vessel occlusion (thrombosis) and inadequate perfusion of vital organs. Either extreme—excessive bleeding or thrombosis—represents a breakdown of the hemostatic mechanism. Common causes of dysregulated hemostasis include hereditary or acquired defects in the clotting mechanism and secondary effects of infection or cancer. The drugs used to inhibit thrombosis and to limit abnormal bleeding are the subjects of this chapter.

MECHANISMS OF BLOOD COAGULATION The vascular endothelial cell layer lining blood vessels has an anticoagulant phenotype, and circulating blood platelets and clotting factors do not normally adhere to it to an appreciable extent. In the setting of vascular injury, the endothelial cell layer rapidly undergoes a series of changes resulting in a more procoagulant phenotype. Injury exposes reactive subendothelial matrix proteins such as collagen and von Willebrand factor, which results in platelet adherence and activation, and secretion and synthesis of vasoconstrictors and plateletrecruiting and activating molecules. Thus, thromboxane A 2 (TXA2 ) is synthesized from arachidonic acid within platelets and is a platelet activator and potent vasoconstrictor. Products secreted from platelet granules include adenosine diphosphate (ADP), a powerful inducer of platelet aggregation, and serotonin (5-HT), which stimulates aggregation and vasoconstriction. Activation of platelets results in a conformational change in the αIIbβIII integrin (IIb/IIIa) receptor, enabling it to bind fibrinogen, which cross-links adjacent platelets, resulting in aggregation and formation of a platelet plug (Figure 34–1). Simultaneously, the coagulation system cascade is activated, resulting in thrombin generation and a fibrin clot, which stabilizes the platelet plug (see below). Knowledge of the hemostatic mechanism is important for diagnosis of bleeding disorders. Patients with defects in the formation of the primary platelet plug (defects in primary hemostasis, eg, platelet function defects, von Willebrand disease) typically bleed from surface sites (gingiva, skin, heavy menses) with injury. In contrast, patients with defects in the clotting mechanism (secondary hemostasis, eg, hemophilia A) tend to bleed into deep tissues (joints, muscle, retroperitoneum), often with no apparent inciting event, and bleeding may recur unpredictably.


FIGURE 34–1 Thrombus formation at the site of the damaged vascular wall (EC, endothelial cell) and the role of platelets and clotting factors. Platelet membrane receptors include the glycoprotein (GP) Ia receptor, binding to collagen (C); GP Ib receptor, binding von Willebrand factor (vWF); and GP IIb/IIIa, which binds fibrinogen and other macromolecules. Antiplatelet prostacyclin (PGI2 ) is released from the endothelium. Aggregating substances released from the degranulating platelet include adenosine diphosphate (ADP), thromboxane A2 (TXA2 ), and serotonin (5-HT). Production of factor Xa by intrinsic and extrinsic pathways is detailed in Figure 34–2. (Redrawn and reproduced, with permission, from Simoons ML, Decker JW: New directions in anticoagulant and antiplatelet treatment. [Editorial.] Br Heart J 1995;74:337.)

The platelet is central to normal hemostasis and thromboembolic disease, and is the target of many therapies discussed in this chapter. Platelet-rich thrombi (white thrombi) form in the high flow rate and high shear force environment of arteries. Occlusive arterial thrombi cause serious disease by producing downstream ischemia of extremities or vital organs, and can result in limb amputation or organ failure. Venous clots tend to be more fibrin-rich, contain large numbers of trapped red blood cells, and are recognized pathologically as red thrombi. Deep venous thrombi (DVT) can cause severe swelling and pain of the affected extremity, but the most feared consequence is pulmonary embolism (PE). This occurs when part or all of the clot breaks off from its location in the deep venous system and travels as an embolus through the right side of the heart and into the pulmonary arterial circulation. Occlusion of a large pulmonary artery by an embolic clot can precipitate acute right heart failure and sudden death. In addition lung ischemia or infarction will occur distal to the occluded pulmonary arterial segment. Such emboli usually arise from the deep venous system of the proximal lower extremities or pelvis. Although all thrombi are mixed, the platelet nidus dominates the arterial thrombus and the fibrin tail dominates the venous thrombus.

BLOOD COAGULATION CASCADE Blood coagulates due to the transformation of soluble fibrinogen into insoluble fibrin by the enzyme thrombin. Several circulating proteins interact in a cascading series of limited proteolytic reactions (Figure 34–2). At each step, a clotting factor zymogen undergoes limited proteolysis and becomes an active protease (eg, factor VII is converted to factor VIIa). Each protease factor activates the next clotting


factor in the sequence, culminating in the formation of thrombin (factor IIa). Several of these factors are targets for drug therapy (Table 34–1). TABLE 34–1 Blood clotting factors and drugs that affect them.1



FIGURE 34–2 A model of blood coagulation. With tissue factor (TF), factor VII forms an activated complex (VIIa-TF) that catalyzes the activation of factor IX to factor IXa. Activated factor XIa also catalyzes this reaction. Tissue factor pathway inhibitor inhibits the catalytic action of the VIIa-TF complex. The cascade proceeds as shown, resulting ultimately in the conversion of fibrinogen to fibrin, an essential component of a functional clot. The two major anticoagulant drugs, heparin and warfarin, have very different actions. Heparin, acting in the blood, directly activates anticlotting factors, specifically antithrombin, which inactivates the factors enclosed in rectangles. Warfarin, acting in the liver, inhibits the synthesis of the factors enclosed in circles. Proteins C and S exert anticlotting effects by inactivating activated factors Va and VIIIa.


Thrombin has a central role in hemostasis and has many functions. In clotting, thrombin proteolytically cleaves small peptides from fibrinogen, allowing fibrinogen to polymerize and form a fibrin clot. Thrombin also activates many upstream clotting factors, leading to more thrombin generation, and activates factor XIII, a transaminase that cross-links the fibrin polymer and stabilizes the clot. Thrombin is a potent platelet activator and mitogen. Thrombin also exerts anticoagulant effects by activating the protein C pathway, which attenuates the clotting response (Figure 34–2). It should therefore be apparent that the response to vascular injury is a complex and precisely modulated process that ensures that under normal circumstances, repair of vascular injury occurs without thrombosis and downstream ischemia; that is, the response is proportionate and reversible. Eventually vascular remodeling and repair occur with reversion to the quiescent resting anticoagulant endothelial cell phenotype.

Initiation of Clotting: The Tissue Factor-VIIa Complex The main initiator of blood coagulation in vivo is the tissue factor (TF)-factor VIIa pathway (Figure 34–2). Tissue factor is a transmembrane protein ubiquitously expressed outside the vasculature, but not normally expressed in an active form within vessels. The exposure of TF on damaged endothelium or to blood that has extravasated into tissue binds TF to factor VIIa. This complex, in turn, activates factors X and IX. Factor Xa along with factor Va forms the prothrombinase complex on activated cell surfaces, which catalyzes the conversion of prothrombin (factor II) to thrombin (factor IIa). Thrombin, in turn, activates upstream clotting factors, primarily factors V, VIII, and XI, resulting in amplification of thrombin generation. The TF-factor VIIa-catalyzed activation of factor Xa is regulated by tissue factor pathway inhibitor (TFPI). Thus after initial activation of factor X to Xa by TF-VIIa, further propagation of the clot is by feedback amplification of thrombin through the intrinsic pathway factors VIII and IX (this provides an explanation of why patients with deficiency of factor VIII or IX—hemophilia A and hemophilia B, respectively—have a severe bleeding disorder). It is also important to note that the coagulation mechanism in vivo does not occur in solution, but is localized to activated cell surfaces expressing anionic phospholipids such as phosphatidylserine, and is mediated by Ca2+ bridging between the anionic phospholipids and γcarboxyglutamic acid residues of the clotting factors. This is the basis for using calcium chelators such as ethylenediamine tetraacetic acid (EDTA) or citrate to prevent blood from clotting in a test tube. Antithrombin (AT) is an endogenous anticoagulant and a member of the serine protease inhibitor (serpin) family; it inactivates the serine proteases IIa, IXa, Xa, XIa, and XIIa. The endogenous anticoagulants protein C and protein S attenuate the blood clotting cascade by proteolysis of the two cofactors Va and VIIIa. From an evolutionary standpoint, it is of interest that factors V and VIII have an identical overall domain structure and considerable homology, consistent with a common ancestor gene; likewise the serine proteases are descendants of a trypsin-like common ancestor. Thus, the TF-VIIa initiating complex, serine proteases, and cofactors each have their own lineage-specific attenuation mechanism (Figure 34–2). Defects in natural anticoagulants result in an increased risk of venous thrombosis. The most common defect in the natural anticoagulant system is a mutation in factor V (factor V Leiden), which results in resistance to inactivation by the protein C, protein S mechanism.

Fibrinolysis Fibrinolysis refers to the process of fibrin digestion by the fibrin-specific protease, plasmin. The fibrinolytic system is similar to the coagulation system in that the precursor form of the serine protease plasmin circulates in an inactive form as plasminogen. In response to injury, endothelial cells synthesize and release tissue plasminogen activator (t-PA), which converts plasminogen to plasmin ( Figure 34–3). Plasmin remodels the thrombus and limits its extension by proteolytic digestion of fibrin.


FIGURE 34–3 Schematic representation of the fibrinolytic system. Plasmin is the active fibrinolytic enzyme. Several clinically useful activators are shown on the left in bold. Anistreplase is a combination of streptokinase and the proactivator plasminogen. Aminocaproic acid (right) inhibits the activation of plasminogen to plasmin and is useful in some bleeding disorders. t-PA, tissue plasminogen activator. Both plasminogen and plasmin have specialized protein domains (kringles) that bind to exposed lysines on the fibrin clot and impart clot specificity to the fibrinolytic process. It should be noted that this clot specificity is only observed at physiologic levels of t-PA. At the pharmacologic levels of t-PA used in thrombolytic therapy, clot specificity is lost and a systemic lytic state is created, with attendant increase in bleeding risk. As in the coagulation cascade, there are negative regulators of fibrinolysis: endothelial cells synthesize and release plasminogen activator inhibitor (PAI), which inhibits t-PA; in addition ι 2 antiplasmin circulates in the blood at high concentrations and under physiologic conditions will rapidly inactivate any plasmin that is not clot-bound. However, this regulatory system is overwhelmed by therapeutic doses of plasminogen activators. If the coagulation and fibrinolytic systems are pathologically activated, the hemostatic system may careen out of control, leading to generalized intravascular clotting and bleeding. This process is called disseminated intravascular coagulation (DIC) and may follow massive tissue injury, advanced cancers, obstetric emergencies such as abruptio placentae or retained products of conception, or bacterial sepsis. The treatment of DIC is to control the underlying disease process; if this is not possible, DIC is often fatal. Regulation of the fibrinolytic system is useful in therapeutics. Increased fibrinolysis is effective therapy for thrombotic disease. Tissue plasminogen activator, urokinase, and streptokinase all activate the fibrinolytic system (Figure 34–3). Conversely, decreased fibrinolysis protects clots from lysis and reduces the bleeding of hemostatic failure. Aminocaproic acid is a clinically useful inhibitor of fibrinolysis. Heparin and the oral anticoagulant drugs do not affect the fibrinolytic mechanism.

BASIC PHARMACOLOGY OF THE ANTICOAGULANT DRUGS The ideal anticoagulant drug would prevent pathologic thrombosis and limit reperfusion injury, yet allow a normal response to vascular injury and limit bleeding. Theoretically this could be accomplished by preservation of the TF-VIIa initiation phase of the clotting mechanism with attenuation of the secondary intrinsic pathway propagation phase of clot development. At this time such a drug does not exist; all anticoagulants and fibrinolytic drugs have an increased bleeding risk as their principle toxicity.


INDIRECT THROMBIN INHIBITORS The indirect thrombin inhibitors are so-named because their antithrombotic effect is exerted by their interaction with a separate protein, antithrombin. Unfractionated heparin (UFH), also known as high-molecular-weight (HMW) heparin, low-molecular-weight (LMW) heparin, and the synthetic pentasaccharide fondaparinux bind to antithrombin and enhance its inactivation of factor Xa (Figure 34–4). Unfractionated heparin and to a lesser extent LMW heparin also enhance antithrombin’s inactivation of thrombin.

FIGURE 34–4 Cartoon illustrating differences between low-molecular-weight (LMW) heparins and high-molecular-weight heparin (unfractionated heparin). Fondaparinux is a small pentasaccharide fragment of heparin. Activated antithrombin III (AT III) degrades thrombin, factor X, and several other factors. Binding of these drugs to AT III can increase the catalytic action of AT III 1000-fold. The combination of AT III with unfractionated heparin increases degradation of both factor Xa and thrombin. Combination with fondaparinux or LMW heparin more selectively increases degradation of Xa.

HEPARIN Chemistry & Mechanism of Action Heparin is a heterogeneous mixture of sulfated mucopolysaccharides. It binds to endothelial cell surfaces and a variety of plasma proteins. Its biologic activity is dependent upon the endogenous anticoagulant antithrombin. Antithrombin inhibits clotting factor proteases, especially thrombin (IIa), IXa, and Xa, by forming equimolar stable complexes with them. In the absence of heparin, these reactions are slow; in the presence of heparin, they are accelerated 1000-fold. Only about a third of the molecules in commercial heparin preparations have an accelerating effect because the remainder lack the unique pentasaccharide sequence needed for high-affinity binding to antithrombin. The active heparin molecules bind tightly to antithrombin and cause a conformational change in this inhibitor. The conformational change of antithrombin exposes its active site for more rapid interaction with the proteases (the activated clotting factors). Heparin functions as a cofactor for the antithrombin-protease reaction without being consumed. Once the antithrombin-protease complex is formed, heparin is released intact for renewed binding to more antithrombin. The antithrombin binding region of commercial unfractionated heparin consists of repeating sulfated disaccharide units composed of D-glucosamine-L-iduronic acid and D-glucosamine-D-glucuronic acid. High-molecular-weight fractions of heparin with high affinity for antithrombin markedly inhibit blood coagulation by inhibiting all three factors, especially thrombin and factor Xa. Unfractionated heparin has a molecular weight range of 5000–30,000. In contrast, the shorter-chain, low-molecular-weight fractions of heparin inhibit activated factor X but have less effect on thrombin than the HMW species. Nevertheless, numerous studies have demonstrated that LMW heparins such as enoxaparin, dalteparin, and tinzaparin are effective in several thromboembolic conditions. In fact, these LMW heparins—in comparison with UFH—have equal efficacy, increased bioavailability from the subcutaneous site of injection, and less frequent dosing requirements (once or twice daily is sufficient). Because commercial heparin consists of a family of molecules of different molecular weights extracted from porcine intestinal mucosa and bovine lung, the correlation between the concentration of a given heparin preparation and its effect on coagulation often is poor. Therefore, UFH is standardized by bioassay. Heparin was reformulated in 2009 in response to heparin contamination events in 2007 and 2008. The contaminant was identified as over-sulfated chondroitin sulfate and linked to more than150 adverse events in patients, most commonly hypotension, nausea, and dyspnea within 30 minutes of infusion. In response to this event, heparin sodium was


reformulated with stricter quality control measures and bioassays to make detection of contaminants easier. This reformulation led to a decrease in potency of approximately 10% from the previous formulation. USP heparin is now harmonized to the World Health Organization International Standard (IS) unit dose. Enoxaparin is obtained from the same sources as regular UFH, but doses are specified in milligrams. Fondaparinux is also specified in milligrams. Dalteparin, tinzaparin, and danaparoid (an LMW heparinoid containing heparan sulfate, dermatan sulfate, and chondroitin sulfate), on the other hand, are specified in anti-factor Xa units.

Monitoring of Heparin Effect Close monitoring of the activated partial thromboplastin time (aPTT or PTT) is necessary in patients receiving UFH. Levels of UFH may also be determined by protamine titration (therapeutic levels 0.2–0.4 unit/mL) or anti-Xa units (therapeutic levels 0.3–0.7 unit/mL). Weight-based dosing of the LMW heparins results in predictable pharmacokinetics and plasma levels in patients with normal renal function. Therefore, LMW heparin levels are not generally measured except in the setting of renal insufficiency, obesity, and pregnancy. LMW heparin levels can be determined by anti-Xa units. For enoxaparin, peak therapeutic levels should be 0.5–1 unit/mL for twice-daily dosing, determined 4 hours after administration, and approximately 1.5 units/mL for once-daily dosing.

Toxicity A. Bleeding and Miscellaneous Effects The major adverse effect of heparin is bleeding. This risk can be decreased by scrupulous patient selection, careful control of dosage, and close monitoring. Elderly women and patients with renal failure are more prone to hemorrhage. Heparin is of animal origin and should be used cautiously in patients with allergy. Increased loss of hair and reversible alopecia have been reported. Long-term heparin therapy is associated with osteoporosis and spontaneous fractures. Heparin accelerates the clearing of postprandial lipemia by causing the release of lipoprotein lipase from tissues, and long-term use is associated with mineralocorticoid deficiency. B. Heparin-Induced Thrombocytopenia Heparin-induced thrombocytopenia (HIT) is a systemic hypercoagulable state that occurs in 1–4% of individuals treated with UFH for a minimum of 7 days. Surgical patients are at greatest risk. The reported incidence of HIT is lower in pediatric populations outside the critical care setting and is relatively rare in pregnant women. The risk of HIT may be higher in individuals treated with UFH of bovine origin compared with porcine heparin and is lower in those treated exclusively with LMW heparin. Morbidity and mortality in HIT are related to thrombotic events. Venous thrombosis occurs most commonly, but occlusion of peripheral or central arteries is not infrequent. If an indwelling catheter is present, the risk of thrombosis is increased in that extremity. Skin necrosis has been described, particularly in individuals treated with warfarin in the absence of a direct thrombin inhibitor, presumably due to acute depletion of the vitamin K-dependent anticoagulant protein C occurring in the presence of high levels of procoagulant proteins and an active hypercoagulable state. The following points should be considered in all patients receiving heparin: Platelet counts should be performed frequently; thrombocytopenia appearing in a time frame consistent with an immune response to heparin should be considered suspicious for HIT; and any new thrombus occurring in a patient receiving heparin therapy should raise suspicion of HIT. Patients who develop HIT are treated by discontinuance of heparin and administration of a direct thrombin inhibitor.

Contraindications Heparin is contraindicated in patients with HIT, hypersensitivity to the drug, active bleeding, hemophilia, significant thrombocytopenia, purpura, severe hypertension, intracranial hemorrhage, infective endocarditis, active tuberculosis, ulcerative lesions of the gastrointestinal tract, threatened abortion, visceral carcinoma, or advanced hepatic or renal disease. Heparin should be avoided in patients who have recently had surgery of the brain, spinal cord, or eye; and in patients who are undergoing lumbar puncture or regional anesthetic block. Despite the apparent lack of placental transfer, heparin should be used in pregnant women only when clearly indicated.

Administration & Dosage The indications for the use of heparin are described in the section on clinical pharmacology. A plasma concentration of heparin of 0.2– 0.4 unit/mL (by protamine titration) or 0.3–0.7 unit/mL (anti-Xa units) is considered to be the therapeutic range for treatment of venous thromboembolic disease. This concentration generally corresponds to a PTT of 1.5–2.5 times baseline. However, the use of the PTT for heparin monitoring is problematic. There is no standardization scheme for the PTT as there is for the prothrombin time (PT) and its international normalized ratio (INR) in warfarin monitoring. The PTT in seconds for a given heparin concentration varies between different reagent/instrument systems. Thus, if the PTT is used for monitoring, the laboratory should determine the clotting time that corresponds to the therapeutic range by protamine titration or anti-Xa activity, as listed above. In addition, some patients have a prolonged baseline PTT due to factor deficiency or inhibitors (which could increase bleeding risk) or


lupus anticoagulant (which is not associated with bleeding risk but may be associated with thrombosis risk). Using the PTT to assess heparin effect in such patients is very difficult. An alternative is to use anti-Xa activity to assess heparin concentration, a test now widely available on automated coagulation instruments. This approach more accurately measures the heparin concentration; however, it does not provide the global assessment of intrinsic pathway integrity of the PTT. The following strategy is recommended: prior to initiating anticoagulant therapy of any type, the integrity of the patient’s hemostatic system should be assessed by a careful history of prior bleeding events, and baseline PT and PTT. If there is a prolonged clotting time, the cause of this (deficiency or inhibitor) should be determined prior to initiating therapy, and treatment goals stratified to a risk-benefit assessment. In high-risk patients measuring both the PTT and anti-Xa activity may be useful. When intermittent heparin administration is used, the aPTT or anti-Xa activity should be measured 6 hours after the administered dose to maintain prolongation of the aPTT to 2–2.5 times that of the control value. However, LMW heparin therapy is the preferred option in this case, as no monitoring is required in most patients. Continuous intravenous administration of heparin is accomplished via an infusion pump. After an initial bolus injection of 80–100 units/kg, a continuous infusion of about 15–22 units/kg/h is required to maintain the anti-Xa activity in the range of 0.3–0.7 units/mL. Low-dose prophylaxis is achieved with subcutaneous administration of heparin, 5000 units every 8–12 hours. Because of the danger of hematoma formation at the injection site, heparin must never be administered intramuscularly. Prophylactic enoxaparin is given subcutaneously in a dosage of 30 mg twice daily or 40 mg once daily. Full-dose enoxaparin therapy is 1 mg/kg subcutaneously every 12 hours. This corresponds to a therapeutic anti-factor Xa level of 0.5–1 unit/mL. Selected patients may be treated with enoxaparin 1.5 mg/kg once a day, with a target anti-Xa level of 1.5 units/mL. The prophylactic dosage of dalteparin is 5000 units subcutaneously once a day; therapeutic dosing is 200 units/kg once a day for venous disease or 120 units/kg every 12 hours for acute coronary syndrome. LMW heparin should be used with caution in patients with renal insufficiency or body weight greater than 150 kg. Measurement of the anti-Xa level is useful to guide dosing in these individuals. The synthetic pentasaccharide molecule fondaparinux avidly binds antithrombin with high specific activity, resulting in efficient inactivation of factor Xa. Fondaparinux has a long half-life of 15 hours, allowing for once-daily dosing by subcutaneous administration. Fondaparinux is effective in the prevention and treatment of venous thromboembolism, and does not appear to cross-react with pathologic HIT antibodies in most individuals.

Reversal of Heparin Action Excessive anticoagulant action of heparin is treated by discontinuance of the drug. If bleeding occurs, administration of a specific antagonist such as protamine sulfate is indicated. Protamine is a highly basic, positively charged peptide that combines with negatively charged heparin as an ion pair to form a stable complex devoid of anticoagulant activity. For every 100 units of heparin remaining in the patient, 1 mg of protamine sulfate is given intravenously; the rate of infusion should not exceed 50 mg in any 10-minute period. Excess protamine must be avoided; it also has an anticoagulant effect. Neutralization of LMW heparin by protamine is incomplete. Limited experience suggests that 1 mg of protamine sulfate may be used to partially neutralize 1 mg of enoxaparin. Protamine will not reverse the activity of fondaparinux. Excess danaparoid can be removed by plasmapheresis.

WARFARIN & OTHER COUMARIN ANTICOAGULANTS Chemistry & Pharmacokinetics The clinical use of the coumarin anticoagulants began with the discovery of an anticoagulant substance formed in spoiled sweet clover silage which caused hemorrhagic disease in cattle. At the behest of local farmers, a chemist at the University of Wisconsin identified the toxic agent as bishydroxycoumarin. Dicumarol, a synthesized derivative, and its congeners, most notably warfarin (Wisconsin Alumni Research Foundation, with “-arin” from coumarin added; Figure 34–5), were initially used as rodenticides. In the 1950s, warfarin (under the brand name Coumadin) was introduced as an antithrombotic agent in humans. Warfarin is one of the most commonly prescribed drugs, used by approximately 1.5 million individuals, and several studies have indicated that the drug is significantly underused in clinical situations where it has proven benefit.


FIGURE 34–5 Structural formulas of several oral anticoagulant drugs and of vitamin K. The carbon atom of warfarin shown at the asterisk is an asymmetric center. Warfarin is generally administered as the sodium salt and has 100% oral bioavailability. Over 99% of racemic warfarin is bound to plasma albumin, which may contribute to its small volume of distribution (the albumin space), its long half-life in plasma (36 hours), and the lack of urinary excretion of unchanged drug. Warfarin used clinically is a racemic mixture composed of equal amounts of two enantiomorphs. The levorotatory S-warfarin is four times more potent than the dextrorotatory R-warfarin. This observation is useful in understanding the stereoselective nature of several drug interactions involving warfarin.

Mechanism of Action Coumarin anticoagulants block the γ-carboxylation of several glutamate residues in prothrombin and factors VII, IX, and X as well as the endogenous anticoagulant proteins C and S (Figure 34–2, Table 34–1). The blockade results in incomplete coagulation factor molecules that are biologically inactive. The protein carboxylation reaction is coupled to the oxidation of vitamin K. The vitamin must then be reduced to reactivate it. Warfarin prevents reductive metabolism of the inactive vitamin K epoxide back to its active hydroquinone form (Figure 34–6). Mutational change of the gene for the responsible enzyme, vitamin K epoxide reductase (VKORC1), can give rise to genetic resistance to warfarin in humans and rodents.


FIGURE 34–6 Vitamin K cycle–metabolic interconversions of vitamin K associated with the synthesis of vitamin K–dependent clotting factors. Vitamin K1 or K2 is activated by reduction to the hydroquinone form (KH2 ). Stepwise oxidation to vitamin K epoxide (KO) is coupled to prothrombin carboxylation by the enzyme carboxylase. The reactivation of vitamin K epoxide is the warfarin-sensitive step (warfarin). The R on the vitamin K molecule represents a 20-carbon phytyl side chain in vitamin K1 and a 30- to 65-carbon polyprenyl side chain in vitamin K2 . There is an 8- to 12-hour delay in the action of warfarin. Its anticoagulant effect results from a balance between partially inhibited synthesis and unaltered degradation of the four vitamin K–dependent clotting factors. The resulting inhibition of coagulation is dependent on their degradation half-lives in the circulation. These half-lives are 6, 24, 40, and 60 hours for factors VII, IX, X, and II, respectively. Importantly, protein C has a short half-life similar to factor VIIa. Thus the immediate effect of warfarin is to deplete the procoagulant factor VII and anticoagulant protein C, which can paradoxically create a transient hypercoagulable state due to residual activity of the longer half-life procoagulants in the face of protein C depletion (see below). For this reason in patients with active hypercoagulable states, such as acute DVT or PE, UFH or LMW heparin is always used to achieve immediate anticoagulation until adequate warfarininduced depletion of the procoagulant clotting factors is achieved. The duration of this overlapping therapy is generally 5–7 days.

Toxicity Warfarin crosses the placenta readily and can cause a hemorrhagic disorder in the fetus. Furthermore, fetal proteins with γcarboxyglutamate residues found in bone and blood may be affected by warfarin; the drug can cause a serious birth defect characterized by abnormal bone formation. Thus, warfarin should never be administered during pregnancy. Cutaneous necrosis with reduced activity of protein C sometimes occurs during the first weeks of therapy in patients who have inherited deficiency of protein C. Rarely, the same process causes frank infarction of the breast, fatty tissues, intestine, and extremities. The pathologic lesion associated with the hemorrhagic infarction is venous thrombosis, consistent with a hypercoagulable state due to warfarin-induced depletion of protein C.

Administration & Dosage Treatment with warfarin should be initiated with standard doses of 5–10 mg. The initial adjustment of the prothrombin time takes about 1 week, which usually results in a maintenance dosage of 5–7 mg/d. The prothrombin time (PT) should be increased to a level representing a reduction of prothrombin activity to 25% of normal and maintained there for long-term therapy. When the activity is less than 20%, the warfarin dosage should be reduced or omitted until the activity rises above 20%. Inherited polymorphisms in 2CYP2C9


a nd VKORC1 have significant effects on warfarin dosing; however algorithms incorporating genomic information to predict initial warfarin dosing were no better than standard clinical algorithms in two of three large randomized trials examining this issue (see Chapter 5). The therapeutic range for oral anticoagulant therapy is defined in terms of an international normalized ratio (INR). The INR is the prothrombin time ratio (patient prothrombin time/mean of normal prothrombin time for lab)ISI, where the ISI exponent refers to the International Sensitivity Index, and is dependent on the specific reagents and instruments used for the determination. The ISI serves to relate measured prothrombin times to a World Health Organization reference standard thromboplastin; thus the prothrombin times performed on different properly calibrated instruments with a variety of thromboplastin reagents should give the same INR results for a given sample. For most reagent and instrument combinations in current use, the ISI is close to 1, making the INR roughly the ratio of the patient prothrombin time to the mean normal prothrombin time. The recommended INR for prophylaxis and treatment of thrombotic disease is 2–3. Patients with some types of artificial heart valves (eg, tilting disk) or other medical conditions increasing thrombotic risk have a recommended range of 2.5–3.5. While a prolonged INR is widely used as an indication of integrity of the coagulation system in liver disease and other disorders, it has been validated only in patients in steady state on chronic warfarin therapy. Occasionally patients exhibit warfarin resistance, defined as progression or recurrence of a thrombotic event while in the therapeutic range. These individuals may have their INR target raised (which is accompanied by an increase in bleeding risk) or be changed to an alternative form of anticoagulation (eg, daily injections of LMW heparin or one of the new oral anticoagulants). Warfarin resistance is most commonly seen in patients with advanced cancers, typically of gastrointestinal origin (Trousseau’s syndrome). A recent study has demonstrated the superiority of LMW heparin over warfarin in preventing recurrent venous thromboembolism in patients with cancer.

Drug Interactions The coumarin anticoagulants often interact with other drugs and with disease states. These interactions can be broadly divided into pharmacokinetic and pharmacodynamic effects (Table 34–2). Pharmacokinetic mechanisms for drug interaction with warfarin mainly involve cytochrome P450 CYP2C9 enzyme induction, enzyme inhibition, and reduced plasma protein binding. Pharmacodynamic mechanisms for interactions with warfarin are synergism (impaired hemostasis, reduced clotting factor synthesis, as in hepatic disease), competitive antagonism (vitamin K), and an altered physiologic control loop for vitamin K (hereditary resistance to oral anticoagulants). TABLE 34–2 Pharmacokinetic and pharmacodynamic drug and body interactions with oral anticoagulants.


The most serious interactions with warfarin are those that increase the anticoagulant effect and the risk of bleeding. The most dangerous of these interactions are the pharmacokinetic interactions with the mostly obsolete pyrazolones phenylbutazone and sulfinpyrazone. These drugs not only augment the hypoprothrombinemia but also inhibit platelet function and may induce peptic ulcer disease (see Chapter 36). The mechanisms for their hypoprothrombinemic interaction are a stereoselective inhibition of oxidative metabolic transformation of S-warfarin (the more potent isomer) and displacement of albumin-bound warfarin, increasing the free fraction. For this and other reasons, neither phenylbutazone nor sulfinpyrazone is in common use in the USA. Metronidazole, fluconazole,


and trimethoprim-sulfamethoxazole also stereoselectively inhibit the metabolic transformation of S-warfarin, whereas amiodarone, disulfiram, and cimetidine inhibit metabolism of both enantiomorphs of warfarin (see Chapter 4). Aspirin, hepatic disease, and hyperthyroidism augment warfarin’s effects—aspirin by its effect on platelet function and the latter two by increasing the turnover rate of clotting factors. The third-generation cephalosporins eliminate the bacteria in the intestinal tract that produce vitamin K and, like warfarin, also directly inhibit vitamin K epoxide reductase. Barbiturates and rifampin cause a marked decrease of the anticoagulant effect by induction of the hepatic enzymes that transform racemic warfarin. Cholestyramine binds warfarin in the intestine and reduces its absorption and bioavailability. Pharmacodynamic reductions of anticoagulant effect occur with increased vitamin K intake (increased synthesis of clotting factors), the diuretics chlorthalidone and spironolactone (clotting factor concentration), hereditary resistance (mutation of vitamin K reactivation cycle molecules), and hypothyroidism (decreased turnover rate of clotting factors). Drugs with no significant effect on anticoagulant therapy include ethanol, phenothiazines, benzodiazepines, acetaminophen, opioids, indomethacin, and most antibiotics.

Reversal of Warfarin Action Excessive anticoagulant effect and bleeding from warfarin can be reversed by stopping the drug and administering oral or parenteral vitamin K1 (phytonadione), fresh-frozen plasma, prothrombin complex concentrates, and recombinant factor VIIa (rFVIIa). A fourfactor concentrate containing factors II, VII, IX, and X was recently approved for use in the US. The disappearance of excessive effect is not correlated with plasma warfarin concentrations but rather with reestablishment of normal activity of the clotting factors. A modest excess of anticoagulant effect without bleeding may require no more than cessation of the drug. The warfarin effect can be rapidly reversed in the setting of severe bleeding with the administration of prothrombin complex or rFVIIa coupled with intravenous vitamin K. It is important to note that due to the long half-life of warfarin, a single dose of vitamin K or rFVIIa may not be sufficient.

ORAL DIRECT FACTOR Xa INHIBITORS Oral Xa inhibitors, including rivaroxaban, apixaban, and edoxaban represent a new class of oral anticoagulant drugs that require no monitoring. Along with oral direct thrombin inhibitors (discussed below) these drugs are having a major impact on antithrombotic pharmacotherapy.

Pharmacology Rivaroxaban, apixaban, and edoxaban inhibit factor Xa, in the final common pathway of clotting (see Figure 34–2). These drugs are given as fixed doses and do not require monitoring. They have a rapid onset of action and shorter half-lives than warfarin. Rivaroxaban has high oral bioavailability when taken with food. Following an oral dose, the peak plasma level is achieved within 2–4 hours; the drug is extensively protein-bound. It is a substrate for the cytochrome P450 system and the P-glycoprotein transporter. Drugs inhibiting both CYP3A4 and P-glycoprotein (eg, ketoconazole) result in increased rivaroxaban effect. One third of the drug is excreted unchanged in the urine and the remainder is metabolized and excreted in the urine and feces. The drug half-life is 5–9 hours in patients aged 20–45 years and is increased in the elderly and in those with impaired renal or hepatic function. Apixaban has an oral bioavailability of 50% and prolonged absorption, resulting in a half-life of 12 hours with repeat dosing. The drug is a substrate of the cytochrome P450 system and P-glycoprotein and is excreted in the urine and feces. Similar to rivaroxaban, drugs inhibiting both CYP3A4 and P-glycoprotein, and impairment of renal or hepatic function result in increased drug effect. Edoxaban is an oral anti-Xa drug in clinical development. Randomized controlled trials versus warfarin for treatment of DVT/PE and for prophylaxis of atrial fibrillation were published in 2013 and showed noninferiority to warfarin for thrombotic events and decreased bleeding events. Based on these data it is likely that edoxaban will soon be FDA-approved for both indications.

Administration & Dosage Rivaroxaban is approved for prevention of embolic stroke in patients with atrial fibrillation without valvular heart disease, prevention of venous thromboembolism following hip or knee surgery, and treatment of venous thromboembolic disease (VTE). The prophylactic dosage is 10 mg orally per day for 35 days for hip replacement or 12 days for knee replacement. For treatment of DVT/PE the dosage is 15 mg twice daily for 3 weeks followed by 20 mg/d. Depending on clinical presentation and risk factors, patients with VTE are treated for 3–6 months; rivaroxaban is also approved for prolonged therapy in selected patients to reduce recurrence risk. Apixaban is approved for prevention of stroke in nonvalvular atrial fibrillation. A recent study demonstrated noninferiority of apixaban compared with standard treatment of VTE with LMW heparin and warfarin. The dosage for atrial fibrillation is 5 mg twice daily. All of these drugs are excreted in part by the kidneys and liver. Therefore use of these agents is not recommended for patients with significant renal or hepatic impairment. In contrast with warfarin, whose effect can be reversed with vitamin K or plasma concentrates, no antidotes exist for direct Xa inhibitors.


DIRECT THROMBIN INHIBITORS The direct thrombin inhibitors (DTIs) exert their anticoagulant effect by directly binding to the active site of thrombin, thereby inhibiting thrombin’s downstream effects. This is in contrast to indirect thrombin inhibitors such as heparin and LMW heparin (see above), which act through antithrombin. Hirudin and bivalirudin are large, bivalent DTIs that bind at the catalytic or active site of thrombin as well as at a substrate recognition site. Argatroban and melagatran are small molecules that bind only at the thrombin active site.

PARENTERAL DIRECT THROMBIN INHIBITORS Leeches have been used for bloodletting since the age of Hippocrates. More recently, surgeons have used medicinal leeches (Hirudo medicinalis) to prevent thrombosis in the fine vessels of reattached digits. Hirudin is a specific, irreversible thrombin inhibitor from leech saliva that for a time was available in recombinant form as lepirudin. Its action is independent of antithrombin, which means it can reach and inactivate fibrin-bound thrombin in thrombi. Lepirudin has little effect on platelets or the bleeding time. Like heparin, it must be administered parenterally and is monitored by the aPTT. Lepirudin was approved by the FDA for use in patients with thrombosis related to heparin-induced thrombocytopenia (HIT). Lepirudin is excreted by the kidney and should be used with great caution in patients with renal insufficiency as no antidote exists. Up to 40% of patients who receive long-term infusions develop an antibody directed against the thrombin-lepirudin complex. These antigen-antibody complexes are not cleared by the kidney and may result in an enhanced anticoagulant effect. Some patients reexposed to the drug developed life-threatening anaphylactic reactions. Lepirudin production was discontinued by the manufacturer in 2012. Bivalirudin, another bivalent inhibitor of thrombin, is administered intravenously, with a rapid onset and offset of action. The drug has a short half-life with clearance that is 20% renal and the remainder metabolic. Bivalirudin also inhibits platelet activation and has been FDA-approved for use in percutaneous coronary angioplasty. Argatroban is a small molecule thrombin inhibitor that is FDA-approved for use in patients with HIT with or without thrombosis and coronary angioplasty in patients with HIT. It, too, has a short half-life, is given by continuous intravenous infusion, and is monitored by aPTT. Its clearance is not affected by renal disease but is dependent on liver function; dose reduction is required in patients with liver disease. Patients on argatroban will demonstrate elevated INRs, rendering the transition to warfarin difficult (ie, the INR will reflect contributions from both warfarin and argatroban). (INR is discussed in detail in the discussion of warfarin administration.) A nomogram is supplied by the manufacturer to assist in this transition.

ORAL DIRECT THROMBIN INHIBITORS Advantages of oral direct thrombin inhibitors include predictable pharmacokinetics and bioavailability, which allow for fixed dosing and predictable anticoagulant response, and make routine coagulation monitoring unnecessary. In addition, these agents do not interact with P450-interacting drugs, and their rapid onset and offset of action allow for immediate anticoagulation, thus avoiding the need for overlap with additional anticoagulant drugs. Dabigatran etexilate mesylate is the first oral direct thrombin inhibitor approved by the FDA. Dabigatran was approved in 2010 to reduce risk of stroke and systemic embolism with nonvalvular atrial fibrillation.

Pharmacology Dabigatran and its metabolites are direct thrombin inhibitors. Following oral administration, dabigatran etexilate mesylate is converted to dabigatran. The oral bioavailability is 3–7% in normal volunteers. The drug is a substrate for the P-glycoprotein efflux pump; however, Pglycoprotein inhibitors or inducers do not have a significant effect on drug clearance. Concomitant use of ketoconazole, amiodarone, quinidine, and clopidogrel increases the effect of dabigatran. The half-life of the drug in normal volunteers is 12–17 hours. Renal impairment results in prolonged drug clearance and may require dose adjustment; the drug should be avoided in patients with severe renal impairment.

Administration & Dosage For prevention of stroke and systemic embolism in nonvalvular atrial fibrillation, 150 mg should be given twice daily to patients with creatinine clearance greater than 30 mL/min. For decreased creatinine clearance of 15–30 mL/min, the dosage is 75 mg twice daily. No monitoring is required. Dabigatran will prolong the PTT and thrombin time, which can be used to estimate drug effect if necessary.

Toxicity As with any anticoagulant drug, the primary toxicity of dabigatran is bleeding. In one study, there was an increase in gastrointestinal


adverse reactions and gastrointestinal bleeding compared with warfarin. There was also a trend toward increased bleeding with dabigatran in patients older than 75 years. There is no antidote for dabigatran. In a drug overdose situation, it is important to maintain renal function or dialyze if necessary. Use of recombinant factor VIIa or prothrombin complex concentrates may be considered as an unproven, off-label use in cases of life-threatening bleeding associated with dabigatran use.

Summary of the Newer Oral Anticoagulant Drugs The new oral direct thrombin inhibitors and oral direct Xa inhibitors have consistently shown equivalent antithrombotic efficacy and lower bleeding rates when compared with traditional warfarin therapy. In addition, these drugs offer the advantages of rapid therapeutic effect, no monitoring requirement, and fewer drug interactions in comparison with warfarin, which has a narrow therapeutic window, is affected by diet and many drugs, and requires monitoring for dosage optimization. However the short half-life of the newer anticoagulants has the important consequence that patient noncompliance will quickly lead to loss of anticoagulant effect and risk of thromboembolism. Additionally no antidote exists at present for patients who present with bleeding, although candidate antidotes are in clinical development. Given the convenience of once- or twice-daily oral dosing, lack of a monitoring requirement, and fewer drug and dietary interactions documented thus far, the new oral anticoagulants are challenging warfarin’s dominance in the prevention and therapy of thrombotic disease.

BASIC PHARMACOLOGY OF THE FIBRINOLYTIC DRUGS Fibrinolytic drugs rapidly lyse thrombi by catalyzing the formation of the serine protease plasmin from its precursor zymogen, plasminogen (Figure 34–3). These drugs create a generalized lytic state when administered intravenously. Thus, both protective hemostatic thrombi and target thromboemboli are broken down. The Box: Thrombolytic Drugs for Acute Myocardial Infarction describes the use of these drugs in one major application.

Pharmacology Streptokinase is a protein (but not an enzyme in itself) synthesized by streptococci that combines with the proactivator plasminogen. This enzymatic complex catalyzes the conversion of inactive plasminogen to active plasmin. Urokinase is a human enzyme synthesized by the kidney that directly converts plasminogen to active plasmin. Plasmin itself cannot be used because naturally occurring inhibitors (antiplasmins) in plasma prevent its effects. However, the absence of inhibitors for urokinase and the streptokinase-proactivator complex permits their use clinically. Plasmin formed inside a thrombus by these activators is protected from plasma antiplasmins, which allows it to lyse the thrombus from within.

Thrombolytic Drugs for Acute Myocardial Infarction The paradigm shift in 1980 on the causation of acute myocardial infarction to acute coronary occlusion by a thrombus created the rationale for thrombolytic therapy of this common lethal disease. At that time—and for the first time—intravenous thrombolytic therapy for acute myocardial infarction in the European Cooperative Study Group trial was found to reduce mortality. Later studies, with thousands of patients in each trial, provided enough statistical power for the 20% reduction in mortality to be considered statistically significant. Although the standard of care in areas with adequate facilities and experience in percutaneous coronary intervention (PCI) now favors catheterization and placement of a stent, thrombolytic therapy is still very important where PCI is not readily available. The proper selection of patients for thrombolytic therapy is critical. The diagnosis of acute myocardial infarction is made clinically and is confirmed by electrocardiography. Patients with ST-segment elevation and bundle branch block on electrocardiography have the best outcomes. All trials to date show the greatest benefit for thrombolytic therapy when it is given early, within 6 hours after symptomatic onset of acute myocardial infarction. Thrombolytic drugs reduce the mortality of acute myocardial infarction. The early and appropriate use of any thrombolytic drug probably transcends possible advantages of a particular drug. Plasminogen can also be activated endogenously by tissue plasminogen activators (t-PAs). These activators preferentially activate plasminogen that is bound to fibrin, which (in theory) confines fibrinolysis to the formed thrombus and avoids systemic activation. Recombinant human t-PA is manufactured as alteplase. Reteplase is another recombinant human t-PA from which several amino acid sequences have been deleted. Tenecteplase is a mutant form of t-PA that has a longer half-life, and it can be given as an intravenous bolus. Reteplase and tenecteplase are as effective as alteplase and have simpler dosing schemes because of their longer half-lives.


Indications & Dosage Administration of fibrinolytic drugs by the intravenous route is indicated in cases of pulmonary embolism with hemodynamic instability, severe deep venous thrombosis such as the superior vena caval syndrome, and ascending thrombophlebitis of the iliofemoral vein with severe lower extremity edema. These drugs are also given intra-arterially, especially for peripheral vascular disease. Thrombolytic therapy in the management of acute myocardial infarction requires careful patient selection, the use of a specific thrombolytic agent, and the benefit of adjuvant therapy. Streptokinase is administered by intravenous infusion of a loading dose of 250,000 units, followed by 100,000 units/h for 24–72 hours. Patients with antistreptococcal antibodies can develop fever, allergic reactions, and therapeutic resistance. Urokinase requires a loading dose of 300,000 units given over 10 minutes and a maintenance dose of 300,000 units/h for 12 hours. Alteplase (t-PA) is given as a 15 mg bolus followed by 0.75 mg/kg (up to 50 mg) over 30 minutes and then 0.5 mg/kg (up to 35 mg) over 60 minutes. Reteplase is given as two 10-unit bolus injections, the second administered 30 minutes after the first injection. Tenecteplase is given as a single intravenous bolus ranging from 30 to 50 mg depending on body weight. Recombinant tPA has also been approved for use in acute ischemic stroke within 3 hours of symptom onset. In patients without hemorrhagic infarct or other contraindications, this therapy has been demonstrated to provide better outcomes in several randomized clinical trials. The recommended dose is 0.9 mg/kg, not to exceed 90 mg, with 10% given as a bolus and the remainder during a 1 hour infusion. Streptokinase has been associated with increased bleeding risk in acute ischemic stroke when given at a dose of 1.5 million units, and its use is not recommended in this setting.

BASIC PHARMACOLOGY OF ANTIPLATELET AGENTS Platelet function is regulated by three categories of substances. The first group consists of agents generated outside the platelet that interact with platelet membrane receptors, eg, catecholamines, collagen, thrombin, and prostacyclin. The second category contains agents generated within the platelet that interact with membrane receptors, eg, ADP, prostaglandin D 2 , prostaglandin E2 , and serotonin. A third group comprises agents generated within the platelet that act within the platelet, eg, prostaglandin endoperoxides and thromboxane A 2 , the cyclic nucleotides cAMP and cGMP, and calcium ion. From this list of agents, several targets for platelet inhibitory drugs have been identified (Figure 34–1): inhibition of prostaglandin synthesis (aspirin), inhibition of ADP-induced platelet aggregation (clopidogrel, prasugrel, ticlopidine), and blockade of glycoprotein IIb/IIIa (GP IIb/IIIa) receptors on platelets (abciximab, tirofiban, and eptifibatide). Dipyridamole and cilostazol are additional antiplatelet drugs.

ASPIRIN The prostaglandin thromboxane A 2 is an arachidonate product that causes platelets to change shape, release their granules, and aggregate (see Chapter 18). Drugs that antagonize this pathway interfere with platelet aggregation in vitro and prolong the bleeding time in vivo. Aspirin is the prototype of this class of drugs. As described in Chapter 18, aspirin inhibits the synthesis of thromboxane A 2 by irreversible acetylation of the enzyme cyclooxygenase. Other salicylates and nonsteroidal anti-inflammatory drugs also inhibit cyclooxygenase but have a shorter duration of inhibitory action because they cannot acetylate cyclooxygenase; that is, their action is reversible. The FDA has approved the use of 325 mg/d aspirin for primary prophylaxis of myocardial infarction but urges caution in this use of aspirin by the general population except when prescribed as an adjunct to risk factor management by smoking cessation and lowering of blood cholesterol and blood pressure. Meta-analysis of many published trials of aspirin and other antiplatelet agents also confirms the value of this intervention in the secondary prevention of vascular events among patients with a history of vascular events.

THIENOPYRIDINES: TICLOPIDINE, CLOPIDOGREL, & PRASUGREL Ticlopidine, clopidogrel, and prasugrel reduce platelet aggregation by inhibiting the ADP pathway of platelets. These drugs irreversibly block the ADP receptor on platelets. Unlike aspirin, these drugs have no effect on prostaglandin metabolism. Use of ticlopidine, clopidogrel, or prasugrel to prevent thrombosis is now considered standard practice in patients undergoing placement of a coronary stent. As the indications and adverse effects of these drugs are different, they will be considered individually. Ticlopidine is approved for prevention of stroke in patients with a history of a transient ischemic attack (TIA) or thrombotic stroke, and in combination with aspirin for prevention of coronary stent thrombosis. Adverse effects of ticlopidine include nausea, dyspepsia, and diarrhea in up to 20% of patients, hemorrhage in 5%, and, most seriously, leukopenia in 1%. The leukopenia is detected by regular monitoring of the white blood cell count during the first 3 months of treatment. Development of thrombotic thrombocytopenic purpura has also been associated with the ingestion of ticlopidine. The dosage of ticlopidine is 250 mg twice daily. Because of the significant side effect profile, the use of ticlopidine for stroke prevention should be restricted to those who are intolerant of or have failed aspirin therapy. Dosages of ticlopidine less than 500 mg/d may be efficacious with fewer adverse effects.


Clopidogrel is approved for patients with unstable angina or non-ST-elevation acute myocardial infarction (NSTEMI) in combination with aspirin; for patients with ST-elevation myocardial infarction (STEMI); or recent myocardial infarction, stroke, or established peripheral arterial disease. For NSTEMI, the dosage is a 300 mg loading dose followed by 75 mg daily of clopidogrel, with a daily aspirin dosage of 75–325 mg. For patients with STEMI, the dosage is 75 mg daily of clopidogrel, in association with aspirin as above; and for recent myocardial infarction, stroke, or peripheral vascular disease, the dosage is 75 mg/d. Clopidogrel has fewer adverse effects than ticlopidine and is rarely associated with neutropenia. Thrombotic thrombocytopenic purpura has been reported. Because of its superior adverse effect profile and dosing requirements, clopidogrel is frequently preferred over ticlopidine. The antithrombotic effects of clopidogrel are dose-dependent; within 5 hours after an oral loading dose of 300 mg, 80% of platelet activity will be inhibited. The maintenance dosage of clopidogrel is 75 mg/d, which achieves maximum platelet inhibition. The duration of the antiplatelet effect is 7–10 days. Clopidogrel is a prodrug that requires activation via the cytochrome P450 enzyme isoform CYP2C19. Depending on the single nucleotide polymorphism (SNP) inheritance pattern in CYP2C19, individuals may be poor metabolizers of clopidogrel, and these patients may be at increased risk of cardiovascular events due to inadequate drug effect. The FDA has recommended CYP2C19 genotyping to identify such patients and advises prescribers to consider alternative therapies in poor metabolizers (see Chapter 5). However, more recent studies have questioned the impact of CYP2C19 metabolizer status on outcomes. Drugs that impair CYP2C19 function, such as omeprazole, should be used with caution. Prasugrel, similar to clopidogrel, is approved for patients with acute coronary syndromes. The drug is given as a 60-mg loading dose and then 10 mg/d in combination with aspirin as outlined for clopidogrel. The Trial to assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition with Prasugrel (TRITON-TIMI38) compared prasugrel with clopidogrel in a randomized, double-blind trial with aspirin and other standard therapies managed with percutaneous coronary interventions. This trial showed a reduction in the primary composite cardiovascular end point (cardiovascular death, nonfatal stroke or nonfatal myocardial infarction) for prasugrel in comparison with clopidogrel. However, the major and minor bleeding risk was increased with prasugrel. Prasugrel is contraindicated in patients with history of TIA or stroke because of increased bleeding risk. In contrast to clopidogrel, cytochrome P450 genotype status is not an important factor in prasugrel pharmacology. Ticagrelor is a new type of ADP inhibitor (cyclopentyltriazolopyrimidine) and is also approved for use in patients with acute coronary syndromes in combination with aspirin. A recent large randomized trial, the Platelet Inhibition and Patient Outcomes (PLATO), compared ticagrelor to clopidogrel in patients with acute coronary syndrome. Although this study demonstrated superiority of ticagrelor in the primary end point of cardiovascular death or stroke, increased noncardiac surgical bleeding was reported.

Aspirin & Clopidogrel Resistance The reported incidence of resistance to these drugs varies greatly, from less than 5% to 75%. In part this tremendous variation in incidence reflects the definition of resistance (recurrent thrombosis while on antiplatelet therapy versus in vitro testing), methods by which drug response is measured, and patient compliance. Several methods for testing aspirin and clopidogrel resistance in vitro are now FDA-approved. However, the incidence of drug resistance varies considerably by testing method. These tests may be useful in selected patients to assess compliance or identify patients at increased risk of recurrent thrombotic events. However, their utility in routine clinical decision making outside of clinical trials remains controversial. A recent randomized prospective trial found no benefit over standard therapy when information obtained from monitoring antiplatelet drug effect was used to alter therapy.

BLOCKADE OF PLATELET GLYCOPROTEIN IIb/IIIa RECEPTORS The platelet GP IIb/IIIa (integrin αIIbβ3) receptor functions as a receptor mainly for fibrinogen and vitronectin but also for fibronectin and von Willebrand factor. Activation of this receptor complex is the final common pathway for platelet aggregation. Ligands for GP IIb/IIIa contain an Arg-Gly-Asp (RGD) sequence motif important for ligand binding, and thus RGD constitutes a therapeutic target. There are approximately 50,000 copies of this complex on the surface of each platelet. Persons lacking this receptor have a bleeding disorder called Glanzmann’s thrombasthenia. The GP IIb/IIIa antagonists are used in patients with acute coronary syndromes. These drugs target the platelet GP IIb/IIIa receptor complex shown in Figure 34–1. Abciximab, a chimeric monoclonal antibody directed against the IIb/IIIa complex including the vitronectin receptor, was the first agent approved in this class of drugs. It has been approved for use in percutaneous coronary intervention and in acute coronary syndromes. Eptifibatide is a cyclic peptide derived from rattlesnake venom that contains a variation of the RGD motif (KGD). Tirofiban is a peptidomimetic inhibitor with the RGD sequence motif. Eptifibatide and tirofiban inhibit ligand binding to the IIb/IIIa receptor by their occupancy of the receptor but do not block the vitronectin receptor. Because of their short halflives, they must be given by continuous infusion. Oral formulations of GP IIb/IIIa antagonists are in various stages of development.

ADDITIONAL ANTIPLATELET-DIRECTED DRUGS Dipyridamole is a vasodilator that also inhibits platelet function by inhibiting adenosine uptake and cGMP phosphodiesterase activity.


Dipyridamole by itself has little or no beneficial effect. Therefore, therapeutic use of this agent is primarily in combination with aspirin to prevent cerebrovascular ischemia. It may also be used in combination with warfarin for primary prophylaxis of thromboemboli in patients with prosthetic heart valves. A combination of dipyridamole complexed with 25 mg of aspirin is now available for secondary prophylaxis of cerebrovascular disease. Cilostazol is a newer phosphodiesterase inhibitor that promotes vasodilation and inhibition of platelet aggregation. Cilostazol is used primarily to treat intermittent claudication.

CLINICAL PHARMACOLOGY OF DRUGS USED TO PREVENT CLOTTING VENOUS THROMBOSIS Risk Factors A. Inherited Disorders The inherited disorders characterized by a tendency to form thrombi (thrombophilia) derive from either quantitative or qualitative abnormalities of the natural anticoagulant system. Deficiencies (loss of function mutations) in the natural anticoagulants antithrombin, protein C, and protein S account for approximately 15% of selected patients with juvenile or recurrent thrombosis and 5–10% of unselected cases of acute venous thrombosis. Additional causes of thrombophilia include gain of function mutations such as the factor V Leiden mutation and the prothrombin 20210 mutation, elevated clotting factor and cofactor levels, and hyperhomocysteinemia that together account for the greater number of hypercoagulable patients. Although loss of function mutations are less common, they are associated with the greatest thrombosis risk. Some patients have multiple inherited risk factors or combinations of inherited and acquired risk factors as discussed below. These individuals are at higher risk for recurrent thrombotic events and are often considered candidates for lifelong therapy. B. Acquired Disease The increased risk of thromboembolism associated with atrial fibrillation and with the placement of mechanical heart valves has long been recognized. Similarly, prolonged bed rest, high-risk surgical procedures, and the presence of cancer are clearly associated with an increased incidence of deep venous thrombosis and embolism. Antiphospholipid antibody syndrome is another important acquired risk factor. Drugs may function as synergistic risk factors in concert with inherited risk factors. For example, women who have the factor V Leiden mutation and take oral contraceptives have a synergistic increase in risk.

Antithrombotic Management A. Prevention Primary prevention of venous thrombosis reduces the incidence of and mortality rate from pulmonary emboli. Heparin and warfarin may be used to prevent venous thrombosis. Subcutaneous administration of low-dose unfractionated heparin, LMW heparin, or fondaparinux provides effective prophylaxis. Warfarin is also effective but requires laboratory monitoring of the prothrombin time. B. Treatment of Established Disease Treatment for established venous thrombosis may be initiated with rivaroxaban alone. Alternatively, patients may be treated with unfractionated or LMW heparin for the first 5–7 days, with an overlap with warfarin. Once therapeutic effects of warfarin have been established, therapy with warfarin is continued for 6 weeks to 6 months or longer, depending on the clinical presentation of the patient. In general, patients who have a provoked event (eg, VTE in the postoperative setting with no other risk factors) would be treated on the shorter end of the spectrum, whereas an individual with recurrent VTE or multiple risk factors might be treated indefinitely. Superficial thrombi confined to the calf veins respond well to short courses of LMW heparin. Warfarin readily crosses the placenta. It can cause hemorrhage at any time during pregnancy as well as developmental defects in the fetus when administered during the first trimester. Therefore, venous thromboembolic disease in pregnant women is generally treated with heparin, best administered by subcutaneous injection.

ARTERIAL THROMBOSIS Activation of platelets is considered an essential process for arterial thrombosis. Thus, treatment with platelet-inhibiting drugs such as aspirin and clopidogrel or ticlopidine is indicated in patients with TIAs and strokes or unstable angina and acute myocardial infarction. As discussed above, prasugrel and ticagrelor are alternatives to clopidogrel for patients with acute coronary syndromes managed with percutaneous coronary interventions. In angina and infarction, these drugs are often used in conjunction with β blockers, calcium channel


blockers, and fibrinolytic drugs.

DRUGS USED IN BLEEDING DISORDERS VITAMIN K Vitamin K confers biologic activity upon prothrombin and factors VII, IX, and X by participating in their postribosomal modification. Vitamin K is a fat-soluble substance found primarily in leafy green vegetables. The dietary requirement is low, because the vitamin is additionally synthesized by bacteria that colonize the human intestine. Two natural forms exist: vitamins K 1 and K2 . Vitamin K 1 (phytonadione; Figure 34–5) is found in food. Vitamin K 2 (menaquinone) is found in human tissues and is synthesized by intestinal bacteria. Vitamins K1 and K2 require bile salts for absorption from the intestinal tract. Vitamin K 1 is available clinically in oral and parenteral forms. Onset of effect is delayed for 6 hours but the effect is complete by 24 hours when treating depression of prothrombin activity by excess warfarin or vitamin K deficiency. Intravenous administration of vitamin K1 should be slow, because rapid infusion can produce dyspnea, chest and back pain, and even death. Vitamin K repletion is best achieved with intravenous or oral administration, because its bioavailability after subcutaneous administration is erratic. Vitamin K 1 is currently administered to all newborns to prevent the hemorrhagic disease of vitamin K deficiency, which is especially common in premature infants. The water-soluble salt of vitamin K3 (menadione) should never be used in therapeutics. It is particularly ineffective in the treatment of warfarin overdosage. Vitamin K deficiency frequently occurs in hospitalized patients in intensive care units because of poor diet, parenteral nutrition, recent surgery, multiple antibiotic therapy, and uremia. Severe hepatic failure results in diminished protein synthesis and a hemorrhagic diathesis that is unresponsive to vitamin K.

PLASMA FRACTIONS Sources & Preparations Deficiencies in plasma coagulation factors can cause bleeding (Table 34–3). Spontaneous bleeding occurs when factor activity is less than 5–10% of normal. Factor VIII deficiency (classic hemophilia, or hemophilia A) and factor IX deficiency (Christmas disease, o r hemophilia B) account for most of the heritable coagulation defects. Concentrated plasma fractions and recombinant protein preparations are available for the treatment of these deficiencies. Administration of plasma-derived, heat- or detergent-treated factor concentrates and recombinant factor concentrates are the standard treatments for bleeding associated with hemophilia. Lyophilized factor VIII concentrates are prepared from large pools of plasma. Transmission of viral diseases such as hepatitis B and C and HIV is reduced or eliminated by pasteurization and by extraction of plasma with solvents and detergents. However, this treatment does not remove other potential causes of transmissible diseases such as prions. For this reason, recombinant clotting factor preparations are recommended whenever possible for factor replacement. The best use of these therapeutic materials requires diagnostic specificity of the deficient factor and quantitation of its activity in plasma. Intermediate purity factor VIII concentrates (as opposed to recombinant or high purity concentrates) contain significant amounts of von Willebrand factor. Humate-P is a factor VIII concentrate that is approved by the FDA for the treatment of bleeding associated with von Willebrand disease. Fresh frozen plasma is used for factor deficiencies for which no recombinant form of the protein is available. A four-factor plasma replacement preparation containing vitamin K–dependent factors II VII, IX, and X is available for rapid reversal of warfarin in bleeding patients. TABLE 34–3 Therapeutic products for the treatment of coagulation disorders 1 .



Clinical Uses Hemophilia A and B patients are given factor VIII and IX replacement, respectively, as prophylaxis to prevent bleeding, and in higher doses to treat bleeding events or to prepare for surgery. Desmopressin acetate increases the factor VIII activity of patients with mild hemophilia A or von Willebrand disease. It can be used in preparation for minor surgery such as tooth extraction without any requirement for infusion of clotting factors if the patient has a documented adequate response. High-dose intranasal desmopressin (see Chapter 17) is available and has been shown to be efficacious and well tolerated by patients. Freeze-dried concentrates of plasma containing prothrombin, factors IX and X, and varied amounts of factor VII (Proplex, etc) are commercially available for treating deficiencies of these factors (Table 34–3). Each unit of factor IX per kilogram of body weight raises its activity in plasma 1.5%. Heparin is often added to inhibit coagulation factors activated by the manufacturing process. However, addition of heparin does not eliminate all thromboembolic risk. Some preparations of factor IX concentrate contain activated clotting factors, which has led to their use in treating patients with inhibitors or antibodies to factor VIII or factor IX. Two products are available expressly for this purpose: Autoplex (with factor VIII correctional activity) and FEIBA (Factor Eight Inhibitor Bypassing Activity). These products are not uniformly successful in arresting hemorrhage, and the factor IX inhibitor titers often rise after treatment with them. Acquired inhibitors of coagulation factors may also be treated with porcine factor VIII (for factor VIII inhibitors) and recombinant activated factor VII. Recombinant activated factor VII (NovoSeven) is being increasingly used to treat coagulopathy associated with liver disease and major blood loss in trauma and surgery. These recombinant and plasma-derived factor concentrates are very expensive, and the indications for them are very precise. Therefore, close consultation with a hematologist knowledgeable in this area is essential. Cryoprecipitate is a plasma protein fraction obtainable from whole blood. It is used to treat deficiencies or qualitative abnormalities of fibrinogen, such as that which occurs with disseminated intravascular coagulation and liver disease. A single unit of cryoprecipitate contains 300 mg of fibrinogen. Cryoprecipitate may also be used for patients with factor VIII deficiency and von Willebrand disease if desmopressin is not indicated and a pathogen-inactivated, recombinant, or plasma-derived product is not available. The concentration of factor VIII and von Willebrand factor in cryoprecipitate is not as great as that found in the concentrated plasma fractions. Moreover, cryoprecipitate is not treated in any manner to decrease the risk of viral exposure. For infusion, the frozen cryoprecipitate unit is thawed and dissolved in a small volume of sterile citrate-saline solution and pooled with other units. Rh-negative women with potential for childbearing should receive only Rhnegative cryoprecipitate because of possible contamination of the product with Rh-positive blood cells.

RECOMBINANT FACTOR VIIa Recombinant factor VIIa is approved for treatment of inherited or acquired hemophilia A or B with inhibitors, treatment of bleeding associated with invasive procedures in congenital or acquired hemophilia, or factor VII deficiency. In the European Union, the drug is also approved for treatment of Glanzmann’s thrombasthenia. Factor VIIa initiates activation of the clotting pathway by activating factor IX and factor X in association with tissue factor (see Figure 34–2). The drug is given by bolus injection. For hemophilia A or B with inhibitors and bleeding, the dosage is 90 mg/kg every 2 hours until hemostasis is achieved, and then continued at 3–6 hour intervals until stable. For congenital factor VII deficiency, the recommended dosage is 15–30 mg/kg every 4–6 hours until hemostasis is achieved. Factor VIIa has been widely used for off-label indications, including bleeding with trauma, surgery, intracerebral hemorrhage, and warfarin toxicity. A major concern of off-label use has been the possibility that thrombotic events may be increased. A recent study examined rates of thromboembolic events in 35 placebo-controlled trials where factor VIIa was administered for nonapproved indications. This study found an increase in arterial, but not venous, thrombotic events, particularly among elderly individuals.

FIBRINOLYTIC INHIBITORS: AMINOCAPROIC ACID Aminocaproic acid (EACA), which is chemically similar to the amino acid lysine, is a synthetic inhibitor of fibrinolysis. It competitively inhibits plasminogen activation (Figure 34–3). It is rapidly absorbed orally and is cleared from the body by the kidney. The usual oral dosage of EACA is 6 g four times a day. When the drug is administered intravenously, a 5 g loading dose should be infused over 30 minutes to avoid hypotension. Tranexamic acid is an analog of aminocaproic acid and has the same properties. It is administered orally with a 15 mg/kg loading dose followed by 30 mg/kg every 6 hours. Clinical uses of EACA are as adjunctive therapy in hemophilia, as therapy for bleeding from fibrinolytic therapy, and as prophylaxis for rebleeding from intracranial aneurysms. Treatment success has also been reported in patients with postsurgical gastrointestinal bleeding and postprostatectomy bleeding and bladder hemorrhage secondary to radiation- and drug-induced cystitis. Adverse effects of the drug include intravascular thrombosis from inhibition of plasminogen activator, hypotension, myopathy, abdominal discomfort, diarrhea, and nasal stuffiness. The drug should not be used in patients with disseminated intravascular coagulation or genitourinary bleeding of the upper tract, eg, kidney and ureters, because of the potential for excessive clotting.


SERINE PROTEASE INHIBITORS: APROTININ Aprotinin is a serine protease inhibitor (serpin) that inhibits fibrinolysis by free plasmin and may have other antihemorrhagic effects as well. It also inhibits the plasmin-streptokinase complex in patients who have received that thrombolytic agent. Aprotinin was shown to reduce bleeding—by as much as 50%—from many types of surgery, especially that involving extracorporeal circulation for open heart procedures and liver transplantation. However, clinical trials and internal data from the manufacturer suggested that use of the drug was associated with an increased risk of renal failure, heart attack, and stroke. A prospective trial was initiated in Canada but halted early because of concerns that use of the drug was associated with increased mortality. The drug was removed from the market in 2007.

PREPARATIONS AVAILABLE


REFERENCES Blood Coagulation & Bleeding Disorders Dahlback B: Advances in understanding pathogenic mechanisms of thrombophilic disorders. Blood 2008;112:19. Mannucci PM, Levi M: Prevention and treatment of major blood loss. N Engl J Med 2007;356:2301.

Drugs Used in Thrombotic Disorders


Bauer KA: Pros and cons of new anticoagulants. Hematology Amer Soc Hematol Educ Program 2013;464. Furei B: Do pharmacogenetics have a role in the dosing of vitamin K antagonists? N Engl J Med 2013;369:2345. Fuster V et al: Guided antithrombotic therapy: Current status and future research direction: Report on a National Heart Lung and Blood Institute working group. Circulation 2012;126:1645. Guyatt GH et al: Executive summary: Antithrombotic therapy and prevention of thrombosis: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (9th Edition). Chest 2012;141(Suppl):7S.

CASE STUDY ANSWER This patient has pulmonary embolism secondary to a deep venous thrombosis (DVT). Options for treating this patient include unfractionated heparin or low-molecular-weight heparin followed by warfarin, with INR goal of 2–3 for 3–6 months; or rivaroxaban alone without monitoring. Several new oral anticoagulants are likely to be approved for this indication in the next few years. Given that the thrombotic event occurred in the setting of oral contraceptive use, the patient should be counseled to use an alternative form of contraception.


CHAPTER

35 Agents Used in Dyslipidemia Mary J. Malloy, MD, & John P. Kane, MD, PhD

CASE STUDY A 42-year-old man with moderately severe coronary artery disease has a body mass index (BMI) of 29, increased abdominal girth, and hypertension that is well controlled. In addition to medicine for hypertension, he is taking 40 mg atorvastatin. Current lipid panel (mg/dL): cholesterol 184, triglycerides 200, LDL-C 110, HDL-C 34, non–HDL-C 150. Lipoprotein(a) (Lp[a]) is twice normal. Fasting glucose is 102 mg/dL, HbA1C is 6%, and fasting insulin is 38 μU/mL. Liver enzymes are normal. Creatine kinase level is mildly elevated. The patient is referred for help with management of his dyslipidemia. You advise dietary measures, exercise, and weight loss. Which additional drugs would help him achieve his lipoprotein treatment goals: LDL-C 50–60 mg/dL; triglycerides < 120 mg/dL; HDL-C > 45 mg/dL; and reduced level of Lp(a)? Would this patient also benefit from a drug to manage insulin resistance? If so, which drug?

Plasma lipids are transported in complexes called lipoproteins. Metabolic disorders that involve elevations in any lipoprotein species are termed hyperlipoproteinemias or hyperlipidemias. Hyperlipemia denotes increased levels of triglycerides. The two major clinical sequelae of hyperlipidemias are acute pancreatitis and atherosclerosis. The former occurs in patients with marked hyperlipemia. Control of triglycerides can prevent recurrent attacks of this life-threatening disease. Atherosclerosis is the leading cause of death for both genders in the USA and other Western countries. Lipoproteins that contain apolipoprotein (apo) B-100 convey lipids into the artery wall. These are low-density (LDL), intermediate-density (IDL), verylow-density (VLDL), and lipoprotein(a) (Lp[a]). Remnant lipoproteins formed during the catabolism of chylomicrons that contain the B-48 protein (apo B-48) can also enter the artery wall, contributing to atherosclerosis. Cellular components in atherosclerotic plaques include foam cells, which are transformed macrophages, and smooth muscle cells filled with cholesteryl esters. These cellular alterations result from endocytosis of modified lipoproteins via at least four species of scavenger receptors. Chemical modification of lipoproteins by free radicals creates ligands for these receptors. The atheroma grows with the accumulation of foam cells, collagen, fibrin, and frequently calcium. Whereas such lesions can slowly occlude coronary vessels, clinical symptoms are more frequently precipitated by rupture of unstable atheromatous plaques, leading to activation of platelets and formation of occlusive thrombi. Although treatment of hyperlipidemia can cause slow physical regression of plaques, the well-documented reduction in acute coronary events that follows vigorous lipid-lowering treatment is attributable chiefly to mitigation of the inflammatory activity of macrophages and is evident within 2–3 months after starting therapy. High-density lipoproteins (HDL) exert several antiatherogenic effects. They participate in retrieval of cholesterol from the artery wall and inhibit the oxidation of atherogenic lipoproteins. Low levels of HDL (hypoalphalipoproteinemia) are an independent risk factor for atherosclerotic disease and thus are a potential target for intervention. Cigarette smoking is a major risk factor for coronary disease. It is associated with reduced levels of HDL, impairment of cholesterol retrieval, cytotoxic effects on the endothelium, increased oxidation of lipoproteins, and stimulation of thrombogenesis. Diabetes, also a major risk factor, is another source of oxidative stress. Normal coronary arteries can dilate in response to ischemia, increasing delivery of oxygen to the myocardium. This process is mediated by nitric oxide, acting on smooth muscle cells of the arterial media. This function is impaired by atherogenic lipoproteins, thus aggravating ischemia. Reducing levels of atherogenic lipoproteins and inhibiting their oxidation restores endothelial function. Because atherogenesis is multifactorial, therapy should be directed toward all modifiable risk factors. Atherogenesis is a dynamic process. Quantitative angiographic trials have demonstrated net regression of plaques during aggressive lipid-lowering therapy. Primary and secondary prevention trials have shown significant reduction in mortality from new coronary events and in all-cause mortality.


PATHOPHYSIOLOGY OF HYPERLIPOPROTEINEMIA NORMAL LIPOPROTEIN METABOLISM Structure Lipoproteins have hydrophobic core regions containing cholesteryl esters and triglycerides surrounded by unesterified cholesterol, phospholipids, and apoproteins. Certain lipoproteins contain very high-molecular-weight B proteins that exist in two forms: B-48, formed in the intestine and found in chylomicrons and their remnants; and B-100, synthesized in liver and found in VLDL, VLDL remnants (IDL), LDL (formed from VLDL), and Lp(a) lipoproteins. HDL consist of at least 20 discrete molecular species containing apolipoprotein A-I (apo A-I). About 100 other proteins are known to be distributed variously among the HDL species.

Synthesis & Catabolism A. Chylomicrons Chylomicrons are formed in the intestine and carry triglycerides of dietary origin, unesterified cholesterol, and cholesteryl esters. They transit the thoracic duct to the bloodstream.

ACRONYMS

Triglycerides are removed in extrahepatic tissues through a pathway shared with VLDL that involves hydrolysis by the lipoprotein lipase (LPL) system. Decrease in particle diameter occurs as triglycerides are depleted. Surface lipids and small apoproteins are transferred to HDL. The resultant chylomicron remnants are taken up by receptor-mediated endocytosis into hepatocytes.


B. Very-Low-Density Lipoproteins VLDL are secreted by liver and export triglycerides to peripheral tissues (Figure 35–1). VLDL triglycerides are hydrolyzed by LPL, yielding free fatty acids for storage in adipose tissue and for oxidation in tissues such as cardiac and skeletal muscle. Depletion of triglycerides produces remnants (IDL), some of which undergo endocytosis directly into hepatocytes. The remainder is converted to LDL by further removal of triglycerides mediated by hepatic lipase. This process explains the “beta shift” phenomenon, the increase of LDL (beta-lipoprotein) in serum as hypertriglyceridemia subsides. Increased levels of LDL can also result from increased secretion of VLDL and from decreased LDL catabolism.

FIGURE 35–1 Metabolism of lipoproteins of hepatic origin. The heavy arrows show the primary pathways. Nascent VLDL are secreted via the Golgi apparatus. They acquire additional apo C lipoproteins and apo E from HDL. Very-low-density lipoproteins (VLDL) are converted to VLDL remnants (IDL) by lipolysis via lipoprotein lipase in the vessels of peripheral tissues. In the process, C apolipoproteins and a portion of the apo E are given back to high-density lipoproteins (HDL). Some of the VLDL remnants are converted to LDL by further loss of triglycerides and loss of apo E. A major pathway for LDL degradation involves the endocytosis of LDL by LDL receptors in the liver and the peripheral tissues, for which apo B-100 is the ligand. Dark color denotes cholesteryl esters; light color denotes triglycerides; the asterisk denotes a functional ligand for LDL receptors; triangles indicate apo E; circles and squares represent C apolipoproteins. FFA, free fatty acid; RER, rough endoplasmic reticulum. (Adapted, with permission, from Kane J, Malloy M: Disorders of


lipoproteins. In: Rosenberg RN et al [editors]: The Molecular and Genetic Basis of Neurological Disease. 2nd ed. Butterworth-Heinemann, 1997.)

C. Low-Density Lipoproteins LDL are catabolized chiefly in hepatocytes and other cells by receptor-mediated endocytosis. Cholesteryl esters from LDL are hydrolyzed, yielding free cholesterol for the synthesis of cell membranes. Cells also obtain cholesterol by synthesis via a pathway involving the formation of mevalonic acid by HMG-CoA reductase. Production of this enzyme and of LDL receptors is transcriptionally regulated by the content of cholesterol in the cell. Normally, about 70% of LDL is removed from plasma by hepatocytes. Even more cholesterol is delivered to the liver via IDL and chylomicrons. Unlike other cells, hepatocytes can eliminate cholesterol by secretion in bile and by conversion to bile acids. D. Lp(a) Lipoprotein Lp(a) lipoprotein is formed from LDL and the (a) protein, linked by a disulfide bridge. The (a) protein is highly homologous with plasminogen but is not activated by tissue plasminogen activator. It occurs in a number of isoforms of different molecular weights. Levels of Lp(a) vary from nil to over 2000 nM/L and are determined chiefly by genetic factors. Lp(a) can be found in atherosclerotic plaques and may also contribute to coronary disease by inhibiting thrombolysis. Levels are elevated in certain inflammatory states. The risk of coronary disease is strongly related to the level of Lp(a). A common variant (I4399M) in the coding region is associated with elevated levels. E. High-Density Lipoproteins The apoproteins of HDL are secreted by the liver and intestine. Much of the lipid comes from the surface monolayers of chylomicrons and VLDL during lipolysis. HDL also acquires cholesterol from peripheral tissues, protecting the cholesterol homeostasis of cells. Free cholesterol is transported from the cell membrane chiefly by a transporter, ABCA1, acquired by a small particle termed prebeta-1 HDL, and then esterified by lecithin:cholesterol acyltransferase (LCAT), leading to the formation of larger HDL species. Cholesterol is also exported from macrophages by the ABCG1 transporter and the docking scavenger receptor, SR-BI, to large HDL particles. The cholesteryl esters are transferred to VLDL, IDL, LDL, and chylomicron remnants with the aid of cholesteryl ester transfer protein (CETP). Much of the cholesteryl ester thus transferred is ultimately delivered to the liver by endocytosis of the acceptor lipoproteins. HDL can also deliver cholesteryl esters directly to the liver via SR-BI that does not cause endocytosis of the lipoproteins. At the population level, HDL-C levels relate inversely to atherosclerosis risk. Among individuals, the capacity to accept exported cholesterol can vary widely at identical levels of HDL-C. The ability of peripheral tissues to export cholesterol via the transporter mechanism and the acceptor capacity of HDL are emerging as major determinants of coronary atherosclerosis.

LIPOPROTEIN DISORDERS Lipoprotein disorders are detected by measuring lipids in serum after a 10-hour fast. Risk of heart disease increases with concentrations of the atherogenic lipoproteins, is inversely related to levels of HDL, and is modified by other risk factors (Table 35–1). Evidence from clinical trials suggests that LDL cholesterol levels of 60 mg/dL may be optimal for patients with coronary disease. Ideally, triglycerides should be below 120 mg/dL. Although LDL-C is still the primary target of treatment, reducing the levels of VLDL and IDL is also important. Calculation of non-HDL cholesterol provides a means of assessing levels of all the lipoproteins in the VLDL to LDL cascade. Differentiation of the disorders requires identification of the lipoproteins involved (Table 35–2). Diagnosis of a primary disorder usually requires further clinical and genetic data as well as ruling out secondary hyperlipidemias (Table 35–3). TABLE 35–1 Current blood lipid guidelines.1


Phenotypes of abnormal lipoprotein distribution are described in this section. Drugs mentioned for use in these conditions are described in the following section on basic and clinical pharmacology.

THE PRIMARY HYPERTRIGLYCERIDEMIAS Hypertriglyceridemia is associated with increased risk of coronary disease. VLDL and IDL have been found in atherosclerotic plaques. These patients tend to have cholesterol-rich VLDL of small-particle diameter and small, dense LDL. Hypertriglyceridemic patients with coronary disease or risk equivalents should be treated aggressively. Patients with triglycerides above 700 mg/dL should be treated to prevent acute pancreatitis because the LPL clearance mechanism is saturated at about this level. Hypertriglyceridemia is an important component of the metabolic syndrome, which also includes low levels of HDL-C, insulin resistance, hypertension, and abdominal obesity. Hyperuricemia is frequently present. Insulin resistance appears to be central to this disorder. Management of these patients frequently requires, in addition to a fibrate, the use of metformin, another agent, or both (see Chapter 41). The severity of hypertriglyceridemia of any cause is increased in the presence of the metabolic syndrome or type 2 diabetes.

Primary Chylomicronemia Chylomicrons are not present in the serum of normal individuals who have fasted 10 hours. The recessive traits of deficiency of LPL or its cofactor, apo C-II, are usually associated with severe lipemia (2000–3000 mg/dL of triglycerides when the patient is consuming a typical American diet). These disorders might not be diagnosed until an attack of acute pancreatitis occurs. Patients may have eruptive


xanthomas, hepatosplenomegaly, hypersplenism, and lipid-laden foam cells in bone marrow, liver, and spleen. The lipemia is aggravated by estrogens because they stimulate VLDL production, and pregnancy may cause marked increases in triglycerides despite strict dietary control. Although these patients have a predominant chylomicronemia, they may also have moderately elevated VLDL, presenting with a pattern called mixed lipemia (fasting chylomicronemia and elevated VLDL). LPL deficiency is diagnosed by assay of lipolytic activity after intravenous injection of heparin. A presumptive diagnosis is made by demonstrating a pronounced decrease in triglycerides a few days after reduction of daily fat intake below 15 g. Marked restriction of total dietary fat is the basis of effective long-term treatment. Niacin, a fibrate, or marine omega-3 fatty acids may be of some benefit if VLDL levels are increased. Genetic variants at other loci that participate in intravascular lipolysis, including LMF1, apo A-V, GPI-HDL BP1, and apo C-III, can have profound effects on triglyceride levels.

Familial Hypertriglyceridemia A. Severe (Usually Mixed Lipemia) Mixed lipemia usually results from impaired removal of triglyceride-rich lipoproteins. Factors that increase VLDL production aggravate the lipemia because VLDL and chylomicrons are competing substrates for LPL. The primary mixed lipemias probably reflect a variety of genetic determinants. Most patients have centripetal obesity with insulin resistance. Other factors that increase secretion of VLDL also worsen the lipemia. Eruptive xanthomas, lipemia retinalis, epigastric pain, and pancreatitis are variably present depending on the severity of the lipemia. Treatment is primarily dietary, with restriction of total fat, avoidance of alcohol and exogenous estrogens, weight reduction, exercise, and supplementation with marine omega-3 fatty acids. Most patients also require treatment with a fibrate. If insulin resistance is not present, niacin may be useful. B. Moderate Increased levels of VLDL can also reflect a genetic predisposition and are worsened by factors that increase the rate of VLDL secretion from liver, ie, obesity, alcohol, diabetes, and estrogens. Treatment includes addressing these issues and the use of fibrates or niacin as needed. Marine omega-3 fatty acids are a valuable adjuvant.

Familial Combined Hyperlipoproteinemia (FCH) In this common disorder associated with an increased incidence of coronary disease, individuals may have elevated levels of VLDL, LDL, or both, and the pattern may change with time. Familial combined hyperlipoproteinemia involves an approximate doubling in VLDL secretion and appears to be transmitted as a dominant trait. Triglycerides can be increased by the factors noted above. Elevations of cholesterol and triglycerides are generally moderate, and xanthomas are absent. Diet alone does not normalize lipid levels. A reductase inhibitor alone, or in combination with niacin or fenofibrate, is usually required to treat these patients. When fenofibrate is combined with a reductase inhibitor, either pravastatin or rosuvastatin is recommended because neither is metabolized via CYP3A4. Marine omega-3 fatty acids may be useful.

Familial Dysbetalipoproteinemia In this disorder, remnants of chylomicrons and VLDL accumulate and levels of LDL are decreased. Because remnants are rich in cholesteryl esters, the level of total cholesterol may be as high as that of triglycerides. Diagnosis is confirmed by the absence of the ε3 and ε4 alleles of apo E, the ε2/ε2 genotype. Other apo E isoforms that lack receptor ligand properties can also be associated with this disorder. Patients often develop tuberous or tuberoeruptive xanthomas, or characteristic planar xanthomas of the palmar creases. They tend to be obese, and some have impaired glucose tolerance. These factors, as well as hypothyroidism, can aggravate the lipemia. Coronary and peripheral atherosclerosis occurs with increased frequency. Weight loss, together with decreased fat, cholesterol, and alcohol consumption, may be sufficient, but a fibrate or niacin is usually needed to control the condition. These agents can be given together in more resistant cases. Reductase inhibitors are also effective because they increase hepatic LDL receptors that participate in remnant removal.

THE PRIMARY HYPERCHOLESTEROLEMIAS Familial Hypercholesterolemia (FH) Familial hypercholesterolemia is an autosomal dominant trait. Although levels of LDL tend to increase throughout childhood, the diagnosis can often be made on the basis of elevated umbilical cord blood cholesterol. In most heterozygotes, cholesterol levels range from 260 to 500 mg/dL. Triglycerides are usually normal. Tendon xanthomas are often present. Arcus corneae and xanthelasma may appear in the third decade. Coronary disease tends to occur prematurely. In homozygous familial hypercholesterolemia, which can lead to coronary disease in childhood, levels of cholesterol often exceed 1000 mg/dL and early tuberous and tendinous xanthomas occur. These patients


may also develop elevated plaque-like xanthomas of the aortic valve, digital webs, buttocks, and extremities. Defects of LDL receptors underlie familial hypercholesterolemia. Some individuals have combined heterozygosity for alleles producing nonfunctional and kinetically impaired receptors. In heterozygous patients, LDL can be normalized with reductase inhibitors or combined drug regimens (Figure 35–2). Homozygotes and those with combined heterozygosity whose receptors retain even minimal function may partially respond to niacin, ezetimibe, and reductase inhibitors. Emerging therapies for these patients include mipomersen, employing an antisense strategy targeted at apo B-100, and lomitapide, a small molecule inhibitor of microsomal triglyceride transfer protein (MTP). LDL apheresis is effective in medication-refractory patients.

FIGURE 35–2 Sites of action of HMG-CoA reductase inhibitors, niacin, ezetimibe, and resins used in treating hyperlipidemias. Lowdensity lipoprotein (LDL) receptors are increased by treatment with resins and HMG-CoA reductase inhibitors. VLDL, very-low-density lipoproteins; R, LDL receptor.

Familial Ligand-Defective Apolipoprotein B-100 Defects in the domain of apo B-100 that binds to the LDL receptor impair the endocytosis of LDL, leading to hypercholesterolemia of moderate severity. Tendon xanthomas may occur. Response to reductase inhibitors is variable. Up-regulation of LDL receptors in liver increases endocytosis of LDL precursors but does not increase uptake of ligand-defective LDL particles. Niacin often has beneficial effects by reducing VLDL production.

Familial Combined Hyperlipoproteinemia (FCH) As described, some persons with familial combined hyperlipoproteinemia have only an elevation in LDL. Serum cholesterol is often less


than 350 mg/dL. Dietary and drug treatment, usually with a reductase inhibitor, is indicated. It may be necessary to add niacin or ezetimibe to normalize LDL.

Lp(a) Hyperlipoproteinemia This familial disorder, which is associated with increased atherogenesis and arterial thrombus formation, is determined chiefly by alleles that dictate increased production of the (a) protein moiety. Lp(a) can be secondarily elevated in patients with severe nephrosis and certain other inflammatory states. Niacin reduces levels of Lp(a) in many patients. Reduction of levels of LDL-C below 100 mg/dL decreases the risk attributable to Lp(a), as does the administration of low dose aspirin.

Cholesteryl Ester Storage Disease Individuals lacking activity of lysosomal acid lipase (LAL), accumulate cholesteryl esters in liver and certain other cell types leading to hepatomegaly with subsequent fibrosis, elevated levels of LDL-C, low levels of HDL-C, and often modest hypertriglyceridemia. Rarely, a totally ablative form, Wolman disease, occurs in infancy. A recombinant replacement enzyme therapy, Sebelipase alfa, is in clinical trials.

Other Disorders Deficiency of cholesterol 7α-hydroxylase can increase LDL in the heterozygous state. Homozygotes can also have elevated triglycerides, resistance to reductase inhibitors, and increased risk of gallstones and coronary disease. Autosomal recessive hypercholesterolemia (ARH) is due to mutations in a protein that normally assists in endocytosis of LDL. The receptor chaperone, PCSK9, normally conducts the receptor to the lysosome for degradation. Gain of function mutations in PCSK9 are associated with elevated levels of LDL-C. The ABCG5 and ABCG8 half-transporters act together in enterocytes and hepatocytes to export phytosterols into the intestinal lumen and bile, respectively. Homozygous or combined heterozygous ablative mutations in either transporter result in elevated levels of LDL enriched in phytosterols, tendon and tuberous xanthomas, and accelerated atherosclerosis. Niacin, ezetimibe, bile acid-binding resins, and reductase inhibitors may be useful, variably, in these disorders.

HDL Deficiency Rare genetic disorders, including Tangier disease and LCAT (lecithin:cholesterol acyltransferase) deficiency, are associated with extremely low levels of HDL. Familial hypoalphalipoproteinemia is a more common disorder with levels of HDL cholesterol usually below 35 mg/dL in men and 45 mg/dL in women. These patients tend to have premature atherosclerosis, and the low HDL may be the only identified risk factor. Management should include special attention to avoidance or treatment of other risk factors. Niacin increases HDL in many of these patients. Reductase inhibitors and fibric acid derivatives exert lesser effects. Aggressive LDL reduction is indicated. In the presence of hypertriglyceridemia, HDL cholesterol is low because of exchange of cholesteryl esters from HDL into triglyceride-rich lipoproteins. Treatment of the hypertriglyceridemia may increase or normalize the HDL level.

SECONDARY HYPERLIPOPROTEINEMIA Before primary disorders can be diagnosed, secondary causes of the phenotype must be considered. The more common conditions are summarized in Table 35–3. The lipoprotein abnormality usually resolves if the underlying disorder can be treated successfully. TABLE 35–3 Secondary causes of hyperlipoproteinemia.


DIETARY MANAGEMENT OF HYPERLIPOPROTEINEMIA Dietary measures are initiated first—unless the patient has evident coronary or peripheral vascular disease—and may obviate the need for drugs. Patients with familial hypercholesterolemia or familial combined hyperlipidemia always require drug therapy. Cholesterol and saturated and trans-fats are the principal factors that increase LDL, whereas total fat, alcohol, and excess calories increase triglycerides. Sucrose and, especially, fructose increase VLDL. Alcohol can cause significant hypertriglyceridemia by increasing hepatic secretion of VLDL. Synthesis and secretion of VLDL are increased by excess calories. During weight loss, LDL and VLDL levels may be much lower than can be maintained during neutral caloric balance. The conclusion that diet suffices for management can be made only after weight has stabilized for at least 1 month. General recommendations include limiting total calories from fat to 20–25% of daily intake, saturated fats to less than 7%, and cholesterol to less than 200 mg/d. Reductions in serum cholesterol range from 10% to 20% on this regimen. Use of complex carbohydrates and fiber is recommended, and cis-monounsaturated fats should predominate. Weight reduction, caloric restriction, and avoidance of alcohol are especially important for patients with elevated VLDL and IDL. The effect of dietary fats on hypertriglyceridemia is dependent on the disposition of double bonds in the fatty acids. Omega-3 fatty acids found in fish oils, but not those from plant sources, activate peroxisome proliferator-activated receptor-alpha (PPAR-α) and can induce profound reduction of triglycerides in some patients. They also have anti-inflammatory and antiarrhythmic activities. Omega-3 fatty acids are available over the counter as triglycerides from marine sources or as a prescription medication (Lovaza) containing ethyl esters of omega-3 fatty acids. The recommended dose of Lovaza is 4 g/d. It is necessary to determine the content of docosahexaenoic


acid and eicosapentaenoic acid in over-the-counter preparations. Appropriate amounts should be taken to provide up to 3–4 g of these fatty acids daily. It is important to select preparations free of mercury and other contaminants. The omega-6 fatty acids present in vegetable oils may cause triglycerides to increase. Patients with primary chylomicronemia and some with mixed lipemia must consume a diet severely restricted in total fat (10–20 g/d, of which 5 g should be vegetable oils rich in essential fatty acids), and fat-soluble vitamins should be given. Homocysteine, which initiates proatherogenic changes in endothelium, can be reduced in many patients by restriction of total protein intake to the amount required for amino acid replacement. Supplementation with folic acid plus other B vitamins, and administration of betaine, a methyl donor, is indicated in severe homocysteinemia. Consumption of red meat should be minimized to reduce the production by the intestinal biome of tetramethyl amine oxide, a compound injurious to arteries.

BASIC & CLINICAL PHARMACOLOGY OF DRUGS USED IN HYPERLIPIDEMIA The decision to use drug therapy for hyperlipidemia is based on the specific metabolic defect and its potential for causing atherosclerosis or pancreatitis. Suggested regimens for the principal lipoprotein disorders are presented in Table 35–2. Diet should be continued to achieve the full potential of the drug regimen. These drugs should be avoided in pregnant and lactating women and those likely to become pregnant. All drugs that alter plasma lipoprotein concentrations potentially require adjustment of doses of warfarin and indandione anticoagulants. Children with heterozygous familial hypercholesterolemia may be treated with a resin or reductase inhibitor, usually after 7 or 8 years of age, when myelination of the central nervous system is essentially complete. The decision to treat a child should be based on the level of LDL, other risk factors, the family history, and the child’s age. Drugs are rarely indicated before age 16 in the absence of multiple risk factors or compound genetic dyslipidemias. TABLE 35–2 The primary hyperlipoproteinemias and their treatment.


COMPETITIVE INHIBITORS OF HMG-COA REDUCTASE (REDUCTASE


INHIBITORS: “STATINS”) These compounds are structural analogs of HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A, Figure 35–3) . Lovastatin, atorvastatin, fluvastatin, pravastatin, simvastatin, rosuvastatin, and pitavastatin belong to this class. They are most effective in reducing LDL. Other effects include decreased oxidative stress and vascular inflammation with increased stability of atherosclerotic lesions. It has become standard practice to initiate reductase inhibitor therapy immediately after acute coronary syndromes, regardless of lipid levels.

FIGURE 35–3 Inhibition of HMG-CoA reductase. Top: The HMG-CoA intermediate that is the immediate precursor of mevalonate, a critical compound in the synthesis of cholesterol. Bottom: The structure of lovastatin and its active form, showing the similarity to the normal HMG-CoA intermediate (shaded areas).

Chemistry & Pharmacokinetics Lovastatin and simvastatin are inactive lactone prodrugs that are hydrolyzed in the gastrointestinal tract to the active β-hydroxyl derivatives, whereas pravastatin has an open, active lactone ring. Atorvastatin, fluvastatin, and rosuvastatin are fluorine-containing congeners that are active as given. Absorption of the ingested doses of the reductase inhibitors varies from 40% to 75% with the exception of fluvastatin, which is almost completely absorbed. All have high first-pass extraction by the liver. Most of the absorbed dose is excreted in the bile; 5–20% is excreted in the urine. Plasma half-lives of these drugs range from 1 to 3 hours except for atorvastatin (14 hours), pitavastatin (12 hours), and rosuvastatin (19 hours).


Mechanism of Action HMG-CoA reductase mediates the first committed step in sterol biosynthesis. The active forms of the reductase inhibitors are structural analogs of the HMG-CoA intermediate (Figure 35–3) that is formed by HMG-CoA reductase in the synthesis of mevalonate. These analogs cause partial inhibition of the enzyme and thus may impair the synthesis of isoprenoids such as ubiquinone and dolichol and the prenylation of proteins. It is not known whether this has biologic significance. However, the reductase inhibitors clearly induce an increase in high-affinity LDL receptors. This effect increases both the fractional catabolic rate of LDL and the liver’s extraction of LDL precursors (VLDL remnants) from the blood, thus reducing LDL ( Figure 35–2). Because of marked first-pass hepatic extraction, the major effect is on the liver. Preferential activity in liver of some congeners appears to be attributable to tissue-specific differences in uptake. Modest decreases in plasma triglycerides and small increases in HDL also occur. Clinical trials involving many of the statins have demonstrated significant reduction of new coronary events and atherothrombotic stroke. Mechanisms other than reduction of lipoprotein levels appear to be involved. The availability of isoprenyl groups from the HMGCoA pathway for prenylation of proteins is reduced by statins, resulting in reduced prenylation of Rho and Rab proteins. Prenylated Rho activates Rho kinase, which mediates a number of mechanisms in vascular biology. The observation that reduction in new coronary events occurs more rapidly than changes in morphology of arterial plaques suggests that these pleiotropic effects may be important. Likewise, decreased prenylation of Rab reduces the accumulation of Aβ protein in neurons, possibly mitigating the manifestations of Alzheimer’s disease. Statins appear to increase the efflux of cholesterol from macrophages, potentially mitigating its accumulation in the artery wall.

Therapeutic Uses & Dosage Reductase inhibitors are useful alone or with resins, niacin, or ezetimibe in reducing levels of LDL. Women with hyperlipidemia who are pregnant, lactating, or likely to become pregnant should not be given these agents. Use in children is restricted to selected patients with familial hypercholesterolemia or familial combined hyperlipidemia. Because cholesterol synthesis occurs predominantly at night, reductase inhibitors—except atorvastatin, rosuvastatin, and pitavastatin —should be given in the evening. Absorption generally (with the exception of pravastatin and pitavastatin) is enhanced by food. Daily doses of lovastatin vary from 10 to 80 mg. Pravastatin is nearly as potent on a mass basis as lovastatin with a maximum recommended daily dose of 80 mg. Simvastatin is twice as potent and is given in doses of 5–80 mg daily. Because of increased risk of myopathy with the 80 mg/d dose, the FDA issued labeling for scaled dosing of simvastatin and Vytorin in 2011. Pitavastatin is given in doses of 1–4 mg daily. Fluvastatin appears to be about half as potent as lovastatin on a mass basis and is given in doses of 10–80 mg daily. Atorvastatin is given in doses of 10–80 mg/d, and rosuvastatin, a very efficacious agent for severe hypercholesterolemia, at 5–40 mg/d. The doseresponse curves of pravastatin and especially of fluvastatin tend to level off in the upper part of the dosage range in patients with moderate to severe hypercholesterolemia. Those of other statins are somewhat more linear.

Toxicity Elevations of serum aminotransferase activity (up to three times normal) occur in some patients. This is often intermittent and usually not associated with other evidence of hepatic toxicity. Therapy may be continued in such patients in the absence of symptoms if aminotransferase levels are monitored and stable. In some patients, who may have underlying liver disease or a history of alcohol abuse, levels may exceed three times normal. This finding portends more severe hepatic toxicity. These patients may present with malaise, anorexia, and precipitous decreases in LDL. Medication should be discontinued immediately in these patients and in asymptomatic patients whose aminotransferase activity is persistently elevated to more than three times the upper limit of normal. These agents should be used with caution and in reduced dosage in patients with hepatic parenchymal disease, north Asians, and the elderly. Severe hepatic disease may preclude their use. In general, aminotransferase activity should be measured at baseline, at 1–2 months, and then every 6– 12 months (if stable). Monitoring of liver enzymes should be more frequent if the patient is taking other drugs that have potential interactions with the statin. Excess intake of alcohol tends to aggravate hepatotoxic effects of statins. Fasting plasma glucose levels tend to increase 5–7 mg/dL with statin treatment. Long-term studies have shown a small but significant increase in the incidence of type 2 diabetes in statin-treated patients, most of whom had findings of prediabetes before treatment. Minor increases in creatine kinase (CK) activity in plasma are observed in some patients receiving reductase inhibitors, frequently associated with heavy physical activity. Rarely, patients may have marked elevations in CK activity, often accompanied by generalized discomfort or weakness in skeletal muscles. If the drug is not discontinued, myoglobinuria can occur, leading to renal injury. Myopathy may occur with monotherapy, but there is an increased incidence in patients also receiving certain other drugs. Genetic variation in an anion transporter (OATP1B1) is associated with severe myopathy and rhabdomyolysis induced by statins. Variants in the gene (SLCO1B1) coding for this protein can now be assessed (see Chapter 5). The catabolism of lovastatin, simvastatin, and atorvastatin proceeds chiefly through CYP3A4, whereas that of fluvastatin and rosuvastatin, and to a lesser extent pitavastatin, is mediated by CYP2C9. Pravastatin is catabolized through other pathways, including sulfation. The 3A4-dependent reductase inhibitors tend to accumulate in plasma in the presence of drugs that inhibit or compete for the 3A4 cytochrome. These include the macrolide antibiotics, cyclosporine, ketoconazole and its congeners, some HIV protease inhibitors,


tacrolimus, nefazodone, fibrates, paroxetine, venlafaxine, and others (see Chapters 4 and 66). Concomitant use of reductase inhibitors with amiodarone or verapamil also causes an increased risk of myopathy. Conversely, drugs such as phenytoin, griseofulvin, barbiturates, rifampin, and thiazolidinediones increase expression of CYP3A4 and can reduce the plasma concentrations of the 3A4-dependent reductase inhibitors. Inhibitors of CYP2C9 such as ketoconazole and its congeners, metronidazole, sulfinpyrazone, amiodarone, and cimetidine may increase plasma levels of fluvastatin and rosuvastatin. Pravastatin and rosuvastatin appear to be the statins of choice for use with verapamil, the ketoconazole group of antifungal agents, macrolides, and cyclosporine. Doses should be kept low and the patient monitored frequently. Plasma levels of lovastatin, simvastatin, and atorvastatin may be elevated in patients ingesting more than 1 liter of grapefruit juice daily. All statins undergo glycosylation, thus creating an interaction with gemfibrozil. Creatine kinase activity should be measured in patients receiving potentially interacting drug combinations. In all patients, CK should be measured at baseline. If muscle pain, tenderness, or weakness appears, CK should be measured immediately and the drug discontinued if activity is elevated significantly over baseline. The myopathy usually reverses promptly upon cessation of therapy. If the association is unclear, the patient can be rechallenged under close surveillance. Myopathy in the absence of elevated CK can occur. Rarely, hypersensitivity syndromes have been reported that include a lupus-like disorder and peripheral neuropathy. Reductase inhibitors may be temporarily discontinued in the event of serious illness, trauma, or major surgery to minimize the potential for liver and muscle toxicity. Use of red yeast rice, a fermentation product that contains statin activity, is not recommended because the statin content is highly variable and some preparations contain a nephrotoxin, citrinin. The long-term safety of these preparations, which often contain a large number of poorly studied organic compounds, has not been established.

FIBRIC ACID DERIVATIVES (FIBRATES) Gemfibrozil and fenofibrate decrease levels of VLDL and, in some patients, LDL as well. Another fibrate, bezafibrate, is not yet available in the USA.

Chemistry & Pharmacokinetics Gemfibrozil is absorbed quantitatively from the intestine and is tightly bound to plasma proteins. It undergoes enterohepatic circulation and readily passes the placenta. The plasma half-life is 1.5 hours. Seventy percent is eliminated through the kidneys, mostly unmodified. The liver modifies some of the drug to hydroxymethyl, carboxyl, or quinol derivatives. Fenofibrate is an isopropyl ester that is hydrolyzed completely in the intestine. Its plasma half-life is 20 hours. Sixty percent is excreted in the urine as the glucuronide, and about 25% in feces.

Mechanism of Action Fibrates function primarily as ligands for the nuclear transcription receptor, PPAR-ι. They transcriptionally up-regulate LPL, apo A-I and apo A-II, and down-regulate apo C-III, an inhibitor of lipolysis. A major effect is an increase in oxidation of fatty acids in liver and striated muscle (Figure 35–4). They increase lipolysis of lipoprotein triglyceride via LPL. Intracellular lipolysis in adipose tissue is


decreased. Levels of VLDL decrease, in part as a result of decreased secretion by the liver. Only modest reductions of LDL occur in most patients. In others, especially those with combined hyperlipidemia, LDL often increases as triglycerides are reduced. HDL cholesterol increases moderately. Part of this apparent increase is a consequence of lower triglyceride in plasma, resulting in reduction in the exchange of triglycerides into HDL in place of cholesteryl esters.

FIGURE 35–4 Hepatic and peripheral effects of fibrates. These effects are mediated by activation of peroxisome proliferator-activated receptor-ι, which modulates the expression of several proteins. LPL, lipoprotein lipase; VLDL, very-low-density lipoproteins.

Therapeutic Uses & Dosage Fibrates are useful drugs in hypertriglyceridemias in which VLDL predominate and in dysbetalipoproteinemia. They also may be of benefit in treating the hypertriglyceridemia that results from treatment with antiviral protease inhibitors. The usual dose of gemfibrozil is 600 mg orally once or twice daily. The dosage of fenofibrate (as Tricor) is one to three 48 mg tablets (or a single 145 mg tablet) daily. Absorption of gemfibrozil is improved when the drug is taken with food.

Toxicity Rare adverse effects of fibrates include rashes, gastrointestinal symptoms, myopathy, arrhythmias, hypokalemia, and high blood levels of aminotransferases or alkaline phosphatase. A few patients show decreases in white blood count or hematocrit. Both agents potentiate the action of coumarin and indanedione anticoagulants, and doses of these agents should be adjusted. Rhabdomyolysis has occurred rarely. Risk of myopathy increases when fibrates are given with reductase inhibitors. Fenofibrate is the fibrate of choice for use in combination with a statin. Fibrates should be avoided in patients with hepatic or renal dysfunction. There appears to be a modest increase in the risk of cholesterol gallstones, reflecting an increase in the cholesterol content of bile. Therefore, fibrates should be used with caution in patients with biliary tract disease or in those at higher risk such as women, obese patients, and Native Americans.


NIACIN (NICOTINIC ACID) Niacin (but not niacinamide) decreases triglycerides and LDL levels, and Lp(a) in most patients. It often increases HDL levels significantly. Historically, combination therapy including niacin has been associated with regression of atherosclerotic coronary lesions in three angiographic trials and with extension of life span in one large trial in which patients received niacin alone. Recently, in a prospective randomized trial, HPS2-THRIVE, 2 grams of extended-release niacin was added to a prostanoid receptor inhibitor (laropiprant) and a statin. No significant reduction of major vascular events was observed in the niacin/laropiprant group vs the group that took the statin alone, but the risk of adverse events was increased. The trial did not adequately address individuals with very high triglycerides or Lp(a), or very low levels of HDL. It is likely that niacin offers therapeutic benefit for such patients and those with statin intolerance.

Chemistry & Pharmacokinetics In its role as a vitamin, niacin (vitamin B3 ) is converted in the body to the amide, which is incorporated into niacinamide adenine dinucleotide (NAD), which in turn has a critical role in energy metabolism. In pharmacologic doses, it has important effects on lipid metabolism that are poorly understood. It is excreted in the urine unmodified and as several metabolites. One, N-methyl nicotinamide, creates a draft on methyl groups that can occasionally result in erythrocyte macrocytosis, similar to deficiency of folate or vitamin B12 .

Mechanism of Action Niacin inhibits VLDL secretion, in turn decreasing production of LDL ( Figure 35–2). Increased clearance of VLDL via the LPL pathway contributes to reduction of triglycerides. Excretion of neutral sterols in the stool is increased acutely as cholesterol is mobilized from tissue pools and a new steady state is reached. The catabolic rate for HDL is decreased. Fibrinogen levels are reduced, and levels of tissue plasminogen activator appear to increase. Niacin inhibits the intracellular lipase of adipose tissue via receptor-mediated signaling, possibly reducing VLDL production by decreasing the flux of free fatty acids to the liver. Sustained inhibition of lipolysis has not been established, however.

Therapeutic Uses & Dosage In combination with a resin or reductase inhibitor, niacin normalizes LDL in most patients with heterozygous familial hypercholesterolemia and other forms of hypercholesterolemia. These combinations are also indicated in some cases of nephrosis. In severe mixed lipemia that is incompletely responsive to diet, niacin often produces marked reduction of triglycerides, an effect enhanced by marine omega-3 fatty acids. It is useful in patients with combined hyperlipidemia and in those with dysbetalipoproteinemia. Niacin is clearly the most effective agent for increasing HDL and the only available agent that may reduce Lp(a). For treatment of heterozygous familial hypercholesterolemia, most patients require 2–6 g of niacin daily; more than this should not be given. For other types of hypercholesterolemia and for hypertriglyceridemia, 1.5–3.5 g daily is often sufficient. Crystalline niacin should be given in divided doses with meals, starting with 100 mg two or three times daily and increasing gradually.

Toxicity Most persons experience a harmless cutaneous vasodilation and sensation of warmth after each dose when niacin is started or the dose increased. Taking 81–325 mg of aspirin one half hour beforehand blunts this prostaglandin-mediated effect. Naproxen, 220 mg once daily, also mitigates the flush. Tachyphylaxis to flushing usually occurs within a few days at doses above 1.5–3 g daily. Patients should be warned to expect the flush and understand that it is a harmless side effect. Pruritus, rashes, dry skin or mucous membranes, and acanthosis nigricans have been reported. The latter requires the discontinuance of niacin because of its association with insulin resistance. Some patients experience nausea and abdominal discomfort. Many can continue the drug at reduced dosage, with inhibitors of gastric acid secretion or with antacids not containing aluminum. Niacin should be avoided in patients with significant peptic disease. Reversible elevations in aminotransferases up to twice normal may occur, usually not associated with liver toxicity. However, liver function should be monitored at baseline and at appropriate intervals. Rarely, true hepatotoxicity may occur, and the drug should be discontinued. The association of severe hepatic dysfunction, including acute necrosis, with the use of over-the-counter sustained-release preparations of niacin has been reported. This effect has not been noted to date with an extended-release preparation, Niaspan, given at bedtime in doses of 2 g or less. Carbohydrate tolerance may be moderately impaired, especially in obese patients, but this is usually reversible except in some patients with latent diabetes. Niacin may be given to diabetics who are receiving insulin and to some receiving oral agents but it may increase insulin resistance. This can often be addressed by increasing the dose of insulin or the oral agents. Hyperuricemia occurs in some patients and occasionally precipitates gout. Allopurinol can be given with niacin if needed. Red cell macrocytosis is not an indication for discontinuing treatment. Significant platelet deficiency can occur rarely and is reversible on cessation of treatment. Rarely, niacin is associated with arrhythmias, mostly atrial, and with macular edema. Patients should be instructed to report blurring of distance vision. Niacin may potentiate the action of antihypertensive agents, requiring adjustment of their dosages. Birth


defects have been reported in offspring of animals given very high doses.

BILE ACID-BINDING RESINS Colestipol, cholestyramine, and colesevelam are useful only for isolated increases in LDL. In patients who also have hypertriglyceridemia, VLDL levels may be further increased during treatment with resins.

Chemistry & Pharmacokinetics The bile acid-binding agents are large polymeric cationic exchange resins that are insoluble in water. They bind bile acids in the intestinal lumen and prevent their reabsorption. The resin itself is not absorbed.

Mechanism of Action Bile acids, metabolites of cholesterol, are normally efficiently reabsorbed in the jejunum and ileum (Figure 35–2). Excretion is increased up to tenfold when resins are given, resulting in enhanced conversion of cholesterol to bile acids in liver via 7α-hydroxylation, which is normally controlled by negative feedback by bile acids. Decreased activation of the FXR receptor by bile acids may result in a modest increase in plasma triglycerides but can also improve glucose metabolism in patients with diabetes. The latter effect is due to increased secretion of the incretin glucagon-like peptide-1 from the intestine, thus increasing insulin secretion. Increased uptake of LDL and IDL from plasma results from up-regulation of LDL receptors, particularly in liver. Therefore, the resins are without effect in patients with homozygous familial hypercholesterolemia who have no functioning receptors but may be useful in those with some residual receptor function and in patients with receptor-defective combined heterozygous states.

Therapeutic Uses & Dosage The resins are used in treatment of patients with primary hypercholesterolemia, producing approximately 20% reduction in LDL cholesterol in maximal dosage. If resins are used to treat LDL elevations in persons with combined hyperlipidemia, they may cause an increase in VLDL, requiring the addition of a second agent such as a fibrate or niacin. Resins are also used in combination with other drugs to achieve further hypocholesterolemic effect (see below). They may be helpful in relieving pruritus in patients who have cholestasis and bile salt accumulation. Because the resins bind digitalis glycosides, they may be useful in digitalis toxicity. Colestipol and cholestyramine are available as granular preparations. A gradual increase of dosage of granules from 4 or 5 g/d to 20 g/d is recommended. Total dosages of 30–32 g/d may be needed for maximum effect. The usual dosage for a child is 10–20 g/d. Granular resins are mixed with juice or water and allowed to hydrate for 1 minute. Colestipol is also available in 1 g tablets that must be swallowed whole, with a maximum dose of 16 g daily. Colesevelam is available in 625 mg tablets and as a suspension (1875 mg or 3750 mg packets). The maximum dose is six tablets or 3750 mg as suspension, daily. Resins should be taken in two or three doses with meals.

Toxicity Common complaints are constipation and bloating, usually relieved by increasing dietary fiber. Resins should be avoided in patients with diverticulitis. Heartburn and diarrhea are occasionally reported. In patients who have preexisting bowel disease or cholestasis, steatorrhea may occur. Malabsorption of vitamin K occurs rarely, leading to hypoprothrombinemia. Prothrombin time should be measured frequently in patients who are taking resins and anticoagulants. Malabsorption of folic acid has been reported rarely. Increased formation of gallstones, particularly in obese persons, was an anticipated adverse effect but has rarely occurred in practice. Absorption of certain drugs, including those with neutral or cationic charge as well as anions, may be impaired by the resins. These include digitalis glycosides, thiazides, warfarin, tetracycline, thyroxine, iron salts, pravastatin, fluvastatin, ezetimibe, folic acid, phenylbutazone, aspirin, and ascorbic acid, among others. In general, additional medication (except niacin) should be given 1 hour before or at least 2 hours after the resin to ensure adequate absorption. Colesevelam does not bind digoxin, warfarin, or reductase inhibitors.

INHIBITORS OF INTESTINAL STEROL ABSORPTION Ezetimibe inhibits intestinal absorption of phytosterols and cholesterol. Its primary clinical effect is reduction of LDL levels. In one trial, patients receiving ezetimibe in combination with simvastatin had marginal, but not statistically significant, increases in carotid intimamedial thickness (IMT) compared with those receiving simvastatin alone. Interpretation of this observation is difficult for several reasons, including the fact that baseline IMT was unexpectedly small, probably due to prior lipid-lowering therapy. Because reducing LDL levels by virtually every modality has been associated with reduced risk of coronary events, it is reasonable to assume that reduction of LDL by ezetimibe will have a similar impact.


Chemistry & Pharmacokinetics Ezetimibe is readily absorbed and conjugated in the intestine to an active glucuronide, reaching peak blood levels in 12–14 hours. It undergoes enterohepatic circulation, and its half-life is 22 hours. Approximately 80% of the drug is excreted in feces. Plasma concentrations are substantially increased when it is administered with fibrates and reduced when it is given with cholestyramine. Other resins may also decrease its absorption. There are no significant interactions with warfarin or digoxin.

Mechanism of Action Ezetimibe selectively inhibits intestinal absorption of cholesterol and phytosterols. A transport protein, NPC1L1, is the target of the drug. It is effective in the absence of dietary cholesterol because it also inhibits reabsorption of cholesterol excreted in the bile.

Therapeutic Uses & Dosage The effect of ezetimibe on cholesterol absorption is constant over the dosage range of 5–20 mg/d. Therefore, a daily dose of 10 mg is used. Average reduction in LDL cholesterol with ezetimibe alone in patients with primary hypercholesterolemia is about 18%, with minimal increases in HDL cholesterol. It is also effective in patients with phytosterolemia. Ezetimibe is synergistic with reductase inhibitors, producing decrements as great as 25% in LDL cholesterol beyond that achieved with the reductase inhibitor alone.

Toxicity Ezetimibe does not appear to be a substrate for cytochrome P450 enzymes. Experience to date reveals a low incidence of reversible impaired hepatic function with a small increase in incidence when given with a reductase inhibitor. Myositis has been reported rarely.

NEWER AGENTS FOR TREATMENT OF DYSLIPIDEMIA INHIBITION OF MICROSOMAL TRIGLYCERIDE TRANSFER PROTEIN Microsomal triglyceride transfer protein (MTP) plays an essential role in the accretion of triglycerides to nascent VLDL in liver, and to chylomicrons in the intestine. Its inhibition decreases VLDL secretion and consequently the accumulation of LDL in plasma. An MTP inhibitor, lomitapide, is available but is currently restricted to patients with homozygous familial hypercholesterolemia. It causes accumulation of triglycerides in the liver in some individuals. Elevations in transaminases can occur. Patients must maintain a low fat diet to avoid steatorrhea but should take steps to minimize deficiency of fat-soluble nutrients. Lomitapide is given orally in gradually increasing doses of 5–60 mg capsules once daily 2 hours after the evening meal. It is available only through a restricted (REMS) program.

ANTISENSE INHIBITION OF APO B-100 SYNTHESIS Mipomersen is an apo B 20-mer antisense oligonucleotide that targets apo B-100, mainly in the liver. It is important to note that the apo B-100 gene is also transcribed in the retina and in cardiomyocytes. Subcutaneous injections of mipomersen reduce levels of LDL and Lp(a). Mild to moderate injection site reactions and flu-like symptoms can occur. The drug is available only for use in homozygous familial hypercholesterolemia through a restricted (REMS) program.

CETP INHIBITION


Cholesteryl ester transfer protein (CETP) inhibitors are under active investigation. The first drug in this class, torcetrapib, aroused great interest because it markedly increased HDL and reduced LDL. However, it was withdrawn from clinical trials because it increased cardiovascular events and deaths in the treatment group. Anacetrapib and evacetrapib are analogs currently in phase 3 clinical trials.

PCSK9 INHIBITION Development of inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) follows on the observation that loss of function mutations result in very low levels of LDL-C and no apparent morbidity. Therapeutic agents currently include antibodies (eg, evolocumab, alirocumab) and antisense oligonucleotides. LDL-C reductions of up to 70% at the highest doses have been achieved with one of these agents when administered parenterally twice weekly. Triglycerides, apo B-100, and Lp(a) were also substantially reduced. No serious adverse effects have been reported in ongoing trials. Development of small molecules with this action is also underway. Studies of this strategy should be approached with caution because of the established role of PCSK9 in normal neuronal apoptosis and cerebral development.

AMP KINASE ACTIVATION AMP-activated protein kinase acts as a sensor of energy status in cells. When increased ATP availability is required, AMP kinase increases fatty acid oxidation and insulin sensitivity, and inhibits cholesterol and triglyceride biosynthesis. Although the trials to date have been directed at decreasing LDL-C levels, AMP kinase activation may have merit for management of the metabolic syndrome and diabetes. An agent combining AMP kinase activation and ATP citrate lyase inhibition is in clinical trials.

TREATMENT WITH DRUG COMBINATIONS Combined drug therapy is useful (1) when VLDL levels are significantly increased during treatment of hypercholesterolemia with a resin; (2) when LDL and VLDL levels are both elevated initially; (3) when LDL or VLDL levels are not normalized with a single agent, or (4) when an elevated level of Lp(a) or an HDL deficiency coexists with other hyperlipidemias. The lowest effective doses should be used in combination therapy and the patient should be monitored more closely for evidence of toxicity.

FIBRIC ACID DERIVATIVES & BILE ACID-BINDING RESINS This combination is sometimes useful in treating patients with familial combined hyperlipidemia who are intolerant of niacin or statins. However, it may increase the risk of cholelithiasis.

HMG-COA REDUCTASE INHIBITORS & BILE ACID-BINDING RESINS This synergistic combination is useful in the treatment of familial hypercholesterolemia but may not control levels of VLDL in some patients with familial combined hyperlipoproteinemia. Statins should be given 1 hour before or at least 2 hours after the resin to ensure their absorption.

NIACIN & BILE ACID-BINDING RESINS This combination effectively controls VLDL levels during resin therapy of familial combined hyperlipoproteinemia or other disorders involving both increased VLDL and LDL levels. When VLDL and LDL levels are both initially increased, doses of niacin as low as 1–3 g/d may be sufficient in combination with a resin. The niacin-resin combination is effective for treating heterozygous familial hypercholesterolemia. The drugs may be taken together, because niacin does not bind to the resins.

NIACIN & REDUCTASE INHIBITORS If the maximum tolerated statin dose fails to achieve the LDL cholesterol goal in a patient with hypercholesterolemia, niacin may be helpful. This combination may be useful in the treatment of familial combined hyperlipoproteinemia.

REDUCTASE INHIBITORS & EZETIMIBE


This combination is highly synergistic in treating primary hypercholesterolemia and has some use in the treatment of patients with homozygous familial hypercholesterolemia who have some receptor function.

REDUCTASE INHIBITORS & FENOFIBRATE Fenofibrate appears to be complementary with most statins in the treatment of familial combined hyperlipoproteinemia and other conditions involving elevations of both LDL and VLDL. The combination of fenofibrate with rosuvastatin appears to be well tolerated. Some other statins may interact unfavorably owing to effects on cytochrome P450 metabolism. In any case, particular vigilance for liver and muscle toxicity is indicated.

COMBINATIONS OF RESINS, EZETIMIBE, NIACIN, & REDUCTASE INHIBITORS These agents act in a complementary fashion to normalize cholesterol in patients with severe disorders involving elevated LDL. The effects are sustained, and little compound toxicity has been observed. Effective doses of the individual drugs may be lower than when each is used alone; for example, as little as 1–2 g of niacin may substantially increase the effects of the other agents.

SUMMARY Drugs Used in Dyslipidemia



PREPARATIONS AVAILABLE

REFERENCES Ballantyne CM et al: Efficacy and safety of a novel dual modulator of adenosine triphosphate-citrate lyase and adenosine monophosphate-activated protein kinase in patients with hypercholesterolemia: Results of a multicenter, randomized, double-blind, placebo-controlled, parallel-group trial. J Am Coll Cardiol 2013;62:1154. Balwani M et al: Clinical effects and safety profile of recombinant human lysosomal acid lipase in patients with cholesteryl ester storage disease. Hepatology 2013;58:950. Boekholdt SM et al: Levels and changes of HDL cholesterol and apolipoprotein A-I in relation to risk of cardiovascular events among statin-treated patients: A metaanalysis. Circulation 2013;128:1504. Bruckert E, Labreuche J, Amarenco P: Meta-analysis of the effect of nicotinic acid alone or in combination on cardiovascular events and atherosclerosis. Atherosclerosis 2010;210:353. Brunzell JD et al: Lipoprotein management in patients with cardiometabolic risk: Consensus conference report from the ADA and the American College of Cardiology Foundation. J Am Coll Cardiol 2008;51(15):1512. Elam M, Lovato E, Ginsberg H: T he role of fibrates in cardiovascular disease prevention, T he ACCORD–lipid perspective. Curr Opin Lipidol 2011;22:55. International Atherosclerosis Society Position Paper: Global Recommendations for the Management of Dyslipidemia. Available at: http://www.athero.org/IASPositionPaper.asp. LaRosa JC et al: Safety and effect of very low levels of low density lipoprotein cholesterol on cardiovascular events. Am J Cardiol 2013;111:1221. Mampuya WM et al: T reatment strategies in patients with statin intolerance: T he Cleveland Clinic experience. Am Heart J 2013;166:597. Perry CM: Lomitapide: A review of its use in adults with homozygous familial hypercholesterolemia. Am J Cardiovasc Drugs 2013;13:265. Ridker PM, Wilson PWF: A trial-based approach to statin guidelines. JAMA 2013; 310:1123. Rosenson RS: AT P III guidelines for treatment of high blood cholesterol. Up to Date 2013. Steinberg D, Grundy SM: T he case for treating hypercholesterolemia at an earlier age: Moving toward consensus. J Am Coll Cardiol 2012;60:2640. Swiger JK et al: Statins and cognition: A systematic review and meta-analysis of short and long term cognitive effects. Mayo Clin Proceed 2013;88:1213. T aylor F et al: Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2013;1:CD004816. Varbo A et al: Remnant cholesterol as a causal risk factor for ischemic heart disease. J Am Coll Cardiol 2013;61:427.

CASE STUDY ANSWER This patient has combined hyperlipidemia. The statin should be continued. A drug that reduces VLDL production would be


beneficial (niacin or fenofibrate). Although niacin is the preferred agent to increase HDL-C and reduce Lp(a), it may increase insulin resistance. The addition of metformin may become necessary. If the LDL-C goal is not reached, the statin dose could be increased or ezetimibe added. Creatine kinase should be monitored. Marine omega-3 fatty acids will help to reduce triglycerides.


CHAPTER

36 Nonsteroidal Anti-Inflammatory Drugs, Disease-Modifying Antirheumatic Drugs, Nonopioid Analgesics, & Drugs Used in Gout Nabeel H. Borazan, MD, & Daniel E. Furst, MD

CASE STUDY A 48-year-old man presents with complaints of bilateral morning stiffness in his wrists and knees and pain in these joints on exercise. On physical examination, the joints are slightly swollen. The rest of the examination is unremarkable. His laboratory findings are also negative except for slight anemia, elevated erythrocyte sedimentation rate, and positive rheumatoid factor. With the diagnosis of rheumatoid arthritis, he is started on a regimen of naproxen, 220 mg twice daily. After 1 week, the dosage is increased to 440 mg twice daily. His symptoms are reduced at this dosage, but he complains of significant heartburn that is not controlled by antacids. He is then switched to celecoxib, 200 mg twice daily, and on this regimen his joint symptoms and heartburn resolve. Two years later, he returns with increased joint symptoms. His hands, wrists, elbows, feet, and knees are all now involved and appear swollen, warm, and tender. What therapeutic options should be considered at this time? What are the possible complications?

ACRONYMS

THE IMMUNE RESPONSE The immune response occurs when immunologically competent cells are activated in response to foreign organisms or antigenic substances liberated during the acute or chronic inflammatory response. The outcome of the immune response for the host may be deleterious if it leads to chronic inflammation without resolution of the underlying injurious process (see Chapter 55). Chronic


inflammation involves the release of multiple cytokines and chemokines plus a very complex interplay of immunoactive cells. The whole range of autoimmune diseases (eg, RA, vasculitis, SLE) and inflammatory conditions (eg, gout) derive from abnormalities in this cascade. The cell damage associated with inflammation acts on cell membranes to release leukocyte lysosomal enzymes; arachidonic acid is then liberated from precursor compounds, and various eicosanoids are synthesized (see Chapter 18). The lipoxygenase pathway of arachidonate metabolism yields leukotrienes, which have a powerful chemotactic effect on eosinophils, neutrophils, and macrophages and promote bronchoconstriction and alterations in vascular permeability. During inflammation, stimulation of the neutrophil membranes produces oxygen-derived free radicals and other reactive molecules such as hydrogen peroxide and hydroxyl radicals. The interaction of these substances with arachidonic acid results in the generation of chemotactic substances, thus perpetuating the inflammatory process.

THERAPEUTIC STRATEGIES The treatment of patients with inflammation involves two primary goals: first, the relief of symptoms and the maintenance of function, which are usually the major continuing complaints of the patient; and second, the slowing or arrest of the tissue-damaging process. In RA, several validated combined indices are used to define response (eg, Disease Activity Index [DAS], American College of Rheumatology Response Index [ACR Response]). These indices often combine joint tenderness and swelling, patient response, and laboratory data. Reduction of inflammation with NSAIDs often results in relief of pain for significant periods. Furthermore, most of the nonopioid analgesics (aspirin, etc) have anti-inflammatory effects, so they are appropriate for the treatment of both acute and chronic inflammatory conditions. The glucocorticoids also have powerful anti-inflammatory effects and when first introduced were considered to be the ultimate answer to the treatment of inflammatory arthritis. Although there are data indicating that low-dose corticosteroids have diseasemodifying properties, their toxicity makes them less favored than other medications, when it is possible to use the others. However, the glucocorticoids continue to have a significant role in the long-term treatment of arthritis. Another important group of agents is characterized as DMARDs including biologics (a subset of the DMARDs). They decrease inflammation, improve symptoms, and slow the bone damage associated with RA. They affect more basic inflammatory mechanisms than do glucocorticoids or the NSAIDs. They may also be more toxic than those alternative medications.

NONSTEROIDAL ANTI-INFLAMMATORY DRUGS Salicylates and other similar agents used to treat rheumatic disease share the capacity to suppress the signs and symptoms of inflammation including pain. These drugs also exert antipyretic effects. Since aspirin, the original NSAID, has a number of adverse effects, many other NSAIDs have been developed in attempts to improve upon aspirin’s efficacy and decrease its toxicity.

Chemistry & Pharmacokinetics The NSAIDs are grouped in several chemical classes, as shown in Figure 36–1. This chemical diversity yields a broad range of pharmacokinetic characteristics (Table 36–1). Although there are many differences in the kinetics of NSAIDs, they have some general properties in common. All but one of the NSAIDs are weak organic acids as given; the exception, nabumetone, is a ketone prodrug that is metabolized to the acidic active drug.


FIGURE 36–1 Chemical structures of some NSAIDs. TABLE 36–1 PROPERTIES OF ASPIRIN AND SOME OTHER NONSTEROIDAL ANTI-INFLAMMATORY DRUGS.



Most of these drugs are well absorbed, and food does not substantially change their bioavailability. Most of the NSAIDs are highly metabolized, some by phase I followed by phase II mechanisms and others by direct glucuronidation (phase II) alone. NSAID metabolism proceeds, in large part, by way of the CYP3A or CYP2C families of P450 enzymes in the liver (see Chapter 4). While renal excretion is the most important route for final elimination, nearly all undergo varying degrees of biliary excretion and reabsorption (enterohepatic circulation). In fact, the degree of lower gastrointestinal (GI) tract irritation correlates with the amount of enterohepatic circulation. Most of the NSAIDs are highly protein-bound (~ 98%), usually to albumin. Most of the NSAIDs (eg, ibuprofen, ketoprofen) are racemic mixtures, while one, naproxen, is provided as a single enantiomer and a few have no chiral center (eg, diclofenac). All NSAIDs can be found in synovial fluid after repeated dosing. Drugs with short half-lives remain in the joints longer than would be predicted from their half-lives, while drugs with longer half-lives disappear from the synovial fluid at a rate proportionate to their halflives.

Pharmacodynamics NSAID anti-inflammatory activity is mediated chiefly through inhibition of prostaglandin biosynthesis (Figure 36–2). Various NSAIDs have additional possible mechanisms of action, including inhibition of chemotaxis, down-regulation of IL-1 production, decreased production of free radicals and superoxide, and interference with calcium-mediated intracellular events. Aspirin irreversibly acetylates and blocks platelet COX, while the non-COX-selective NSAIDs are reversible inhibitors.


FIGURE 36–2 Prostanoid mediators derived from arachidonic acid and sites of drug action. ASA, acetylsalicylic acid (aspirin); LT, leukotriene; NSAID, nonsteroidal anti-inflammatory drug. Selectivity for COX-1 versus COX-2 is variable and incomplete for the older NSAIDs, but selective COX-2 inhibitors have been synthesized. The selective COX-2 inhibitors do not affect platelet function at their usual doses. The efficacy of COX-2-selective drugs equals that of the older NSAIDs, while GI safety may be improved. On the other hand, selective COX-2 inhibitors increase the incidence of edema, hypertension, and possibly, myocardial infarction. As of August 2011, celecoxib and the less selective meloxicam were the only COX-2 inhibitors marketed in the USA. Celecoxib has an FDA-initiated “black box” warning concerning cardiovascular risks. It has been recommended that all NSAID product labels be revised to mention cardiovascular risks. The NSAIDs decrease the sensitivity of vessels to bradykinin and histamine, affect lymphokine production from T lymphocytes, and reverse the vasodilation of inflammation. To varying degrees, all newer NSAIDs are analgesic, anti-inflammatory, and antipyretic, and all (except the COX-2-selective agents and the nonacetylated salicylates) inhibit platelet aggregation. NSAIDs are all gastric irritants and can be associated with GI ulcers and bleeds as well, although as a group the newer agents tend to cause less GI irritation than aspirin.


Nephrotoxicity, reported for all NSAIDs, is due, in part, to interference with the autoregulation of renal blood flow, which is modulated by prostaglandins. Hepatotoxicity can also occur with any NSAID. Although these drugs effectively inhibit inflammation, there is no evidence that—in contrast to drugs such as methotrexate, biologics, and other DMARDs—they alter the course of any arthritic disorder. Several NSAIDs (including aspirin) reduce the incidence of colon cancer when taken chronically. Several large epidemiologic studies have shown a 50% reduction in relative risk for this neoplasm when the drugs are taken for 5 years or longer. The mechanism for this protective effect is unclear. Although not all NSAIDs are approved by the FDA for the whole range of rheumatic diseases, most are probably effective in RA, sero-negative spondyloarthropathies (eg, PA and arthritis associated with inflammatory bowel disease), OA, localized musculoskeletal syndromes (eg, sprains and strains, low back pain), and gout (except tolmetin, which appears to be ineffective in gout). Adverse effects are generally quite similar for all of the NSAIDs: 1. 2. 3. 4. 5. 6. 7. 8.

Central nervous system: Headaches, tinnitus, dizziness, and rarely, aseptic meningitis. Cardiovascular: Fluid retention, hypertension, edema, and rarely, myocardial infarction and congestive heart failure (CHF). Gastrointestinal: Abdominal pain, dysplasia, nausea, vomiting, and rarely, ulcers or bleeding. Hematologic: Rare thrombocytopenia, neutropenia, or even aplastic anemia. Hepatic: Abnormal liver function test results and rare liver failure. Pulmonary: Asthma. Skin: Rashes, all types, pruritus. Renal: Renal insufficiency, renal failure, hyperkalemia, and proteinuria.

ASPIRIN Aspirin’s long use and availability without prescription diminishes its glamour compared with that of the newer NSAIDs. Aspirin is now rarely used as an anti-inflammatory medication and will be reviewed only in terms of its antiplatelet effects (ie, doses of 81–325 mg once daily). 1. Pharmacokinetics: Salicylic acid is a simple organic acid with a pKa of 3.0. Aspirin (acetylsalicylic acid; ASA) has a pKa of 3.5 (see Table 1–3). Aspirin is absorbed as such and is rapidly hydrolyzed (serum half-life 15 minutes) to acetic acid and salicylate by esterases in tissue and blood (Figure 36–3). Salicylate is nonlinearly bound to albumin. Alkalinization of the urine increases the rate of excretion of free salicylate and its water-soluble conjugates.


FIGURE 36–3 Structure and metabolism of the salicylates. (Reproduced, with permission, from Meyers FH, Jawetz E, Goldfien A: Review of Medical Pharmacology, 7th ed. McGraw-Hill, 1980. Copyright © The McGraw-Hill Companies, Inc.) 2. Mechanisms of Action: Aspirin irreversibly inhibits platelet COX so that aspirin’s antiplatelet effect lasts 8–10 days (the life of the platelet). In other tissues, synthesis of new COX replaces the inactivated enzyme so that ordinary doses have a duration of action of 6–12 hours. 3. Clinical Uses: Aspirin decreases the incidence of transient ischemic attacks, unstable angina, coronary artery thrombosis with myocardial infarction, and thrombosis after coronary artery bypass grafting (see Chapter 34). 4. Epidemiologic studies suggest that long-term use of aspirin at low dosage is associated with a lower incidence of colon cancer, possibly related to its COX-inhibiting effects. 5. Adverse Effects: In addition to the common side effects listed above, aspirin’s main adverse effects at antithrombotic doses are gastric upset (intolerance) and gastric and duodenal ulcers. Hepatotoxicity, asthma, rashes, GI bleeding, and renal toxicity rarely if ever occur at antithrombotic doses. 6. The antiplatelet action of aspirin contraindicates its use by patients with hemophilia. Although previously not recommended during pregnancy, aspirin may be valuable in treating preeclampsia-eclampsia.

NONACETYLATED SALICYLATES These drugs include magnesium choline salicylate, sodium salicylate, and salicyl salicylate. All nonacetylated salicylates are effective anti-inflammatory drugs, although they may be less effective analgesics than aspirin. Because they are much less effective than aspirin as COX inhibitors and they do not inhibit platelet aggregation, they may be preferable when COX inhibition is undesirable such as in patients with asthma, those with bleeding tendencies, and even (under close supervision) those with renal dysfunction. The nonacetylated salicylates are administered in doses up to 3–4 g of salicylate a day and can be monitored using serum salicylate measurements.


COX-2 SELECTIVE INHIBITORS COX-2 selective inhibitors, or coxibs, were developed in an attempt to inhibit prostaglandin synthesis by the COX-2 isozyme induced at sites of inflammation without affecting the action of the constitutively active “housekeeping” COX-1 isozyme found in the GI tract, kidneys, and platelets. COX-2 inhibitors at usual doses have no impact on platelet aggregation, which is mediated by thromboxane produced by the COX-1 isozyme. In contrast, they do inhibit COX-2-mediated prostacyclin synthesis in the vascular endothelium. As a result, COX-2 inhibitors do not offer the cardioprotective effects of traditional nonselective NSAIDs. Recommended doses of COX-2 inhibitors cause renal toxicities similar to those associated with traditional NSAIDs. Clinical data suggested a higher incidence of cardiovascular thrombotic events associated with COX-2 inhibitors such as rofecoxib and valdecoxib, resulting in their withdrawal from the market.

Celecoxib Celecoxib is a selective COX-2 inhibitor—about 10–20 times more selective for COX-2 than for COX-1. Pharmacokinetic and dosage considerations are given in Table 36–1. Celecoxib is associated with fewer endoscopic ulcers than most other NSAIDs. Probably because it is a sulfonamide, celecoxib may cause rashes. It does not affect platelet aggregation at usual doses. It interacts occasionally with warfarin—as would be expected of a drug metabolized via CYP2C9. Adverse effects are the common toxicities listed above.

Meloxicam Meloxicam is an enolcarboxamide related to piroxicam that preferentially inhibits COX-2 over COX-1, particularly at its lowest therapeutic dose of 7.5 mg/d. It is not as selective as celecoxib and may be considered “preferentially” selective rather than “highly” selective. It is associated with fewer clinical GI symptoms and complications than piroxicam, diclofenac, and naproxen. Similarly, while meloxicam is known to inhibit synthesis of thromboxane A 2 , even at supratherapeutic doses, its blockade of thromboxane A 2 does not reach levels that result in decreased in vivo platelet function (see common adverse effects above).

NONSELECTIVE COX INHIBITORS* Diclofenac Diclofenac is a phenylacetic acid derivative that is relatively nonselective as a COX inhibitor. Pharmacokinetic and dosage characteristics are set forth in Table 36–1. Gastrointestinal ulceration may occur less frequently than with some other NSAIDs. A preparation combining diclofenac and misoprostol decreases upper gastrointestinal ulceration but may result in diarrhea. Another combination of diclofenac and omeprazole was also effective with respect to the prevention of recurrent bleeding, but renal adverse effects were common in high-risk patients. Diclofenac, 150 mg/d, appears to impair renal blood flow and glomerular filtration rate. Elevation of serum aminotransferases occurs more commonly with this drug than with other NSAIDs. A 0.1% ophthalmic preparation is promoted for prevention of postoperative ophthalmic inflammation and can be used after intraocular lens implantation and strabismus surgery. A topical gel containing 3% diclofenac is effective for solar keratoses. Diclofenac in


rectal suppository form can be considered for preemptive analgesia and postoperative nausea. In Europe, diclofenac is also available as an oral mouthwash and for intramuscular administration.

Diflunisal Although diflunisal is derived from salicylic acid, it is not metabolized to salicylic acid or salicylate. It undergoes an enterohepatic cycle with reabsorption of its glucuronide metabolite followed by cleavage of the glucuronide to again release the active moiety. Diflunisal is subject to capacity-limited metabolism, with serum half-lives at various dosages approximating that of salicylates (Table 36–1). In RA the recommended dose is 500–1000 mg daily in two divided doses. It is claimed to be particularly effective for cancer pain with bone metastases and for pain control in dental (third molar) surgery. A 2% diflunisal oral ointment is a clinically useful analgesic for painful oral lesions. Because its clearance depends on renal function as well as hepatic metabolism, diflunisal’s dosage should be limited in patients with significant renal impairment.

Etodolac Etodolac is a racemic acetic acid derivative with an intermediate half-life (Table 36–1). The analgesic dosage of etodolac is 200–400 mg three to four times daily. The recommended dose in OA and RA is 300 mg twice or three times a day up to 500 mg twice a day initially followed by a maintenance of 600 mg/d.

Flurbiprofen Flurbiprofen is a propionic acid derivative with a possibly more complex mechanism of action than other NSAIDs. Its (S)(−) enantiomer inhibits COX nonselectively, but it has been shown in rat tissue to also affect tumor necrosis factor-α (TNF-α) and nitric oxide synthesis. Hepatic metabolism is extensive; its (R)(+) and (S)(−) enantiomers are metabolized differently, and it does not undergo chiral conversion. It does demonstrate enterohepatic circulation. Flurbiprofen is also available in a topical ophthalmic formulation for inhibition of intraoperative miosis. Flurbiprofen intravenously is effective for perioperative analgesia in minor ear, neck, and nose surgery and in lozenge form for sore throat. Although its adverse effect profile is similar to that of other NSAIDs in most ways, flurbiprofen is also rarely associated with cogwheel rigidity, ataxia, tremor, and myoclonus.

Ibuprofen Ibuprofen is a simple derivative of phenylpropionic acid (Figure 36–1). In doses of about 2400 mg daily, ibuprofen is equivalent to 4 g of aspirin in anti-inflammatory effect. Pharmacokinetic characteristics are given in Table 36–1. Oral ibuprofen is often prescribed in lower doses (<2400 mg/d), at which it has analgesic but not anti-inflammatory efficacy. It is available over the counter in low-dose forms under several trade names. Ibuprofen oral and IV is effective in closing patent ductus arteriosus in preterm infants, with much the same efficacy and safety as indomethacin. A topical cream preparation appears to be absorbed into fascia and muscle; ibuprofen cream was more effective than placebo cream in the treatment of primary knee OA. A liquid gel preparation of ibuprofen, 400 mg, provides prompt relief and good overall efficacy in postsurgical dental pain. In comparison with indomethacin, ibuprofen decreases urine output less and also causes less fluid retention. The drug is relatively contraindicated in individuals with nasal polyps, angio-edema, and bronchospastic reactivity to aspirin. Aseptic meningitis (particularly in patients with SLE), and fluid retention have been reported. The concomitant administration of ibuprofen and aspirin antagonizes the irreversible platelet inhibition induced by aspirin. Thus, treatment with ibuprofen in patients with increased cardiovascular risk may limit the cardioprotective effects of aspirin. Furthermore, the use of ibuprofen concomitantly with aspirin may decrease the total antiinflammatory effect. Common adverse effects are listed on pages 620-621; rare hematologic effects include agranulocytosis and aplastic anemia.

Indomethacin Indomethacin, introduced in 1963, is an indole derivative (Figure 36–1). It is a potent nonselective COX inhibitor and may also inhibit phospholipase A and C, reduce neutrophil migration, and decrease T-cell and B-cell proliferation. Indomethacin differs somewhat from other NSAIDs in its indications and toxicities. It has been used to accelerate closure of patent ductus arteriosus. Indomethacin has been tried in numerous small or uncontrolled trials for many other conditions, including Sweet’s syndrome, juvenile RA, pleurisy, nephrotic syndrome, diabetes insipidus, urticarial vasculitis, postepisiotomy pain, and prophylaxis of heterotopic ossification in arthroplasty.


An ophthalmic preparation is efficacious for conjunctival inflammation and to reduce pain after traumatic corneal abrasion. Gingival inflammation is reduced after administration of indomethacin oral rinse. Epidural injections produce a degree of pain relief similar to that achieved with methylprednisolone in postlaminectomy syndrome. At usual doses, indomethacin has the common side effects listed above. The GI effects may include pancreatitis. Headache is experienced by 15–25% of patients and may be associated with dizziness, confusion, and depression. Renal papillary necrosis has also been observed. A number of interactions with other drugs have been reported (see Chapter 66).

Ketoprofen Ketoprofen is a propionic acid derivative that inhibits both COX (nonselectively) and lipoxygenase. Its pharmacokinetic characteristics are given in Table 36–1. Concurrent administration of probenecid elevates ketoprofen levels and prolongs its plasma half-life. The effectiveness of ketoprofen at dosages of 100–300 mg/d is equivalent to that of other NSAIDs. Its major adverse effects are on the GI tract and the central nervous system (see common adverse effects above).

Nabumetone Nabumetone is the only nonacid NSAID in current use; it is given as a ketone prodrug (Figure 36–1) and resembles naproxen in structure. Its half-life of more than 24 hours (Table 36–1) permits once-daily dosing, and the drug does not appear to undergo enterohepatic circulation. Renal impairment results in a doubling of its half-life and a 30% increase in the area under the curve. Its properties are very similar to those of other NSAIDs, though it may be less damaging to the stomach. Unfortunately, higher dosages (eg, 1500–2000 mg/d) are often needed, and this is a very expensive NSAID. Like naproxen, nabumetone has been associated with pseudoporphyria and photosensitivity in some patients.

Naproxen Naproxen is a naphthylpropionic acid derivative. It is the only NSAID presently marketed as a single enantiomer. Naproxen’s free fraction is significantly higher in women than in men, but half-life is similar in both sexes (Table 36–1). Naproxen is effective for the usual rheumatologic indications and is available in a slow-release formulation, as an oral suspension, and over the counter. A topical preparation and an ophthalmic solution are also available. The incidence of upper GI bleeding in over-the-counter use is low but still double that of over-the-counter ibuprofen (perhaps due to a dose effect). Rare cases of allergic pneumonitis, leukocytoclastic vasculitis, and pseudoporphyria as well as the common NSAIDassociated adverse effects have been noted.

Oxaprozin Oxaprozin is another propionic acid derivative NSAID. As noted in Table 36–1, its major difference from the other members of this subgroup is a very long half-life (50–60 hours), although oxaprozin does not undergo enterohepatic circulation. It is mildly uricosuric. Otherwise, the drug has the same benefits and risks that are associated with other NSAIDs.

Piroxicam Piroxicam, an oxicam (Figure 36–1), is a nonselective COX inhibitor that at high concentrations also inhibits polymorphonuclear leukocyte migration, decreases oxygen radical production, and inhibits lymphocyte function. Its long half-life (Table 36–1) permits once-daily dosing. Piroxicam can be used for the usual rheumatic indications. When piroxicam is used in dosages higher than 20 mg/d, an increased incidence of peptic ulcer and bleeding is encountered—as much as 9.5 times higher than with other NSAIDs (see common adverse effects above).

Sulindac Sulindac is a sulfoxide prodrug. It is reversibly metabolized to the active sulfide metabolite, which is excreted in bile and then reabsorbed from the intestine. The enterohepatic cycling prolongs the duration of action to 12–16 hours. In addition to its rheumatic disease indications, sulindac suppresses familial intestinal polyposis and it may inhibit the development of colon, breast, and prostate cancer in humans. Among the more severe adverse reactions, Stevens-Johnson epidermal necrolysis syndrome, thrombocytopenia, agranulocytosis, and nephrotic syndrome have all been observed. It is sometimes associated with cholestatic liver damage.


Tolmetin Tolmetin is a nonselective COX inhibitor with a short half-life (1–2 hours) and is not often used. It is ineffective (for unknown reasons) in the treatment of gout.

Other NSAIDs Azapropazone, carprofen, meclofenamate, and tenoxicam are rarely used and are not reviewed here.

CHOICE OF NSAID All NSAIDs, including aspirin, are about equally efficacious with a few exceptions—tolmetin seems not to be effective for gout, and aspirin is less effective than other NSAIDs (eg, indomethacin) for AS. Thus, NSAIDs tend to be differentiated on the basis of toxicity and cost-effectiveness. For example, the GI and renal side effects of ketorolac limit its use. Some surveys suggest that indomethacin and tolmetin are the NSAIDs associated with the greatest toxicity, while salsalate, aspirin, and ibuprofen are least toxic. The selective COX-2 inhibitors were not included in these analyses. For patients with renal insufficiency, nonacetylated salicylates may be best. Diclofenac and sulindac are associated with more liver function test abnormalities than other NSAIDs. The relatively expensive, selective COX-2 inhibitor celecoxib is probably safest for patients at high risk for GI bleeding but may have a higher risk of cardiovascular toxicity. Celecoxib or a nonselective NSAID plus omeprazole or misoprostol may be appropriate in patients at highest risk for GI bleeding; in this subpopulation of patients, they are costeffective despite their high acquisition costs. The choice of an NSAID thus requires a balance of efficacy, cost-effectiveness, safety, and numerous personal factors (eg, other drugs also being used, concurrent illness, compliance, medical insurance coverage), so that there is no best NSAID for all patients. There may, however, be one or two best NSAIDs for a specific person.

DISEASE-MODIFYING ANTIRHEUMATIC DRUGS RA is a progressive immunologic disease that causes significant systemic effects, shortens life, and reduces mobility and quality of life. Interest has centered on finding treatments that might arrest—or at least slow—this progression by modifying the disease itself. The effects of disease-modifying therapies may take 2 weeks to 6 months to become clinically evident. These therapies include nonbiologic and biologic disease-modifying antirheumatic drugs (usually designated “DMARDs”). The nonbiologic agents include small molecule drugs such as methotrexate, azathioprine, chloroquine and hydroxychloroquine, cyclophosphamide, cyclosporine, leflunomide, mycophenolate mofetil, and sulfasalazine. Tofacitinib, though marketed as a biologic, is actually a well-tolerated nonbiologic DMARD. Gold salts, which were once extensively used, are no longer recommended because of their significant toxicities and questionable efficacy. Biologics are large-molecule therapeutic agents, usually proteins, that are often produced by recombinant DNA technology. The biologic DMARDs approved for RA include: a T-cell-modulating biologic (abatacept), a B-cell cytotoxic agent (rituximab), an anti-IL-6 receptor antibody (tocilizumab), IL-1-inhibiting agents (anakinra, rilonacept, canakinumab), and the TNF-α-blocking agents (five drugs). The small-molecule DMARDs and biologics are discussed alphabetically, independent of origin.

ABATACEPT 1. Mechanism of action: Abatacept is a co-stimulation modulator biologic that inhibits the activation of T cells (see also Chapter 55). After a T cell has engaged an antigen-presenting cell (APC), a second signal is produced by CD28 on the T cell that interacts with CD80 or CD86 on the APC, leading to T-cell activation. Abatacept (which contains the endogenous ligand CTLA-4) binds to CD80 and 86, thereby inhibiting the binding to CD28 and preventing the activation of T cells. 2. Pharmacokinetics: The recommended dose of abatacept for the treatment of adult patients with RA is three intravenous infusion “induction” doses (day 0, week 2, and week 4), followed by monthly infusions. The dose is based on body weight; patients weighing less than 60 kg receiving 500 mg, those 60–100 kg receiving 750 mg, and those more than 100 kg receiving 1000 mg. Abatacept is also available as a subcutaneous formulation and is given as 125 mg subcutaneously once weekly. JIA can also be treated with abatacept with an induction schedule at day 0, week 2, and week 4, followed by intravenous infusion every 4 weeks. The recommended dose for patients 6–17 years of age and weighing less than 75 kg is 10 mg/kg, while those weighing 75 kg or more follow the adult intravenous doses to a maximum not to exceed 1000 mg. The terminal serum half-life is 13– 16 days. Co-administration with methotrexate, NSAIDs, and corticosteroids does not influence abatacept clearance. Most patients respond to abatacept within 12–16 weeks after the initiation of the treatment; however, some patients can respond


in as few as 2–4 weeks. A study showed equivalence between adalimumab and abatacept. 3. Indications: Abatacept can be used as monotherapy or in combination with methotrexate or other DMARDs in patients with moderate to severe RA or severe PJIA. It is being tested in early RA and methotrexate-naïve patients. 4. Adverse Effects: There is a slightly increased risk of infection (as with other biologic DMARDs), predominantly of the upper respiratory tract. Concomitant use with TNF-α antagonists or other biologics is not recommended due to the increased incidence of serious infection. All patients should be screened for latent tuberculosis and viral hepatitis before starting this medication. Live vaccines should be avoided in patients while taking abatacept and up to 3 months after discontinuation. Infusion-related reactions and hypersensitivity reactions, including anaphylaxis, have been reported but are rare. Anti-abatacept antibody formation is infrequent (<5%) and has no effect on clinical outcomes. There is a possible increase in lymphomas but not other malignancies when using abatacept.

AZATHIOPRINE 1. Mechanism of Action: Azathioprine is a synthetic nonbiologic DMARD that acts through its major metabolite, 6-thioguanine. 6Thioguanine suppresses inosinic acid synthesis, B-cell and T-cell function, immunoglobulin production, and IL-2 secretion (see Chapter 55). 2. Pharmacokinetics: Azathioprine can be given orally or parenterally. Its metabolism is bimodal in humans, with rapid metabolizers clearing the drug four times faster than slow metabolizers. Production of 6-thioguanine is dependent on thiopurine methyltransferase (TPMT), and patients with low or absent TPMT activity (0.3% of the population) are at particularly high risk of myelosuppression by excess concentrations of the parent drug, if dosage is not adjusted. 3. Indications: Azathioprine is approved for use in RA at a dosage of 2 mg/kg/d. It is also used for the prevention of kidney transplant rejection in combination with other immune suppressants. Controlled trials show efficacy in PA, reactive arthritis, polymyositis, SLE, maintenance of remission in vasculitis, and Behçet’s disease. Azathioprine is also used in scleroderma; however, in one study, it was found to be less effective than cyclophosphamide in controlling the progression of scleroderma lung disease. 4. Adverse Effects: Azathioprine’s toxicity includes bone marrow suppression, GI disturbances, and some increase in infection risk. As noted in Chapter 55, lymphomas may be increased with azathioprine use. Rarely, fever, rash, and hepatotoxicity signal acute allergic reactions.

CHLOROQUINE & HYDROXYCHLOROQUINE 1. Mechanism of Action: Chloroquine and hydroxychloroquine are nonbiologic drugs mainly used for malaria (see Chapter 52) and in the rheumatic diseases. The following mechanisms have been proposed: suppression of T-lymphocyte responses to mitogens, inhibition of leukocyte chemotaxis, stabilization of lysosomal enzymes, processing through the Fc-receptor, inhibition of DNA and RNA synthesis, and the trapping of free radicals. 2. Pharmacokinetics: Antimalarials are rapidly absorbed and 50% protein-bound in the plasma. They are very extensively tissuebound, particularly in melanin-containing tissues such as the eyes. The drugs are deaminated in the liver and have blood elimination half-lives of up to 45 days. 3. Indications: Antimalarials are approved for RA, but they are not considered very effective DMARDs. Dose-loading may increase rate of response. There is no evidence that these compounds alter bony damage in RA at their usual dosages (up to 6.4 mg/kg/d for hydroxychloroquine or 200 mg/d for chloroquine). It usually takes 3–6 months to obtain a response. Antimalarials are used very commonly in SLE because they decrease mortality and the skin manifestations, serositis, and joint pains of this disease. They have also been used in Sjögren’s syndrome. 4. Adverse Effects: Although ocular toxicity (see Chapter 52) may occur at dosages greater than 250 mg/d for chloroquine and greater than 6.4 mg/kg/d for hydroxychloroquine, it rarely occurs at lower doses. Nevertheless, ophthalmologic monitoring every 12 months is advised. Other toxicities include dyspepsia, nausea, vomiting, abdominal pain, rashes, and nightmares. These drugs appear to be relatively safe in pregnancy.

CYCLOPHOSPHAMIDE 1. Mechanism of Action: Cyclophosphamide is a synthetic nonbiologic DMARD. Its major active metabolite is phosphoramide mustard, which cross-links DNA to prevent cell replication. It suppresses T-cell and B-cell function by 30–40%; T-cell suppression correlates with clinical response in the rheumatic diseases. Its pharmacokinetics and toxicities are discussed in Chapter 54. 2. Indications: Cyclophosphamide is used regularly at 2 mg/kg/d to treat SLE, vasculitis, Wegener’s granulomatosis, and other severe


rheumatic diseases.

CYCLOSPORINE 1. Mechanism of Action: Cyclosporine is a peptide antibiotic but is considered a nonbiologic DMARD. Through regulation of gene transcription, it inhibits IL-1 and IL-2 receptor production and secondarily inhibits macrophage-T-cell interaction and T-cell responsiveness (see Chapter 55). T-cell-dependent B-cell function is also affected. 2. Pharmacokinetics: Cyclosporine absorption is incomplete and somewhat erratic, although a microemulsion formulation improves its consistency and provides 20–30% bioavailability. Grapefruit juice increases cyclosporine bioavailability by as much as 62%. Cyclosporine is metabolized by CYP3A and consequently is subject to a large number of drug interactions (see Chapters 55 and 66). 3. Indications: Cyclosporine is approved for use in RA and retards the appearance of new bony erosions. Its usual dosage is 3–5 mg/kg/d divided into two doses. Anecdotal reports suggest that it may be useful in SLE, polymyositis and dermatomyositis, Wegener’s granulomatosis, and juvenile chronic arthritis. 4. Adverse Effects: Leukopenia, thrombocytopenia, and to a lesser extent, anemia are predictable. High doses can be cardiotoxic and sterility may occur after chronic dosing at anti-rheumatic doses, especially in women. Bladder cancer is very rare but must be looked for, even 5 years after cessation of use.

LEFLUNOMIDE 1. Mechanism of Action: Leflunomide, another nonbiologic DMARD, undergoes rapid conversion, both in the intestine and in the plasma, to its active metabolite, A77-1726. This metabolite inhibits dihydroorotate dehydrogenase, leading to a decrease in ribonucleotide synthesis and the arrest of stimulated cells in the G1 phase of cell growth. Consequently, leflunomide inhibits T-cell proliferation and reduces production of autoantibodies by B cells. Secondary effects include increases of IL-10 receptor mRNA, decreased IL-8 receptor type A mRNA, and decreased TNF-α-dependent nuclear factor kappa B (NF-κB) activation. 2. Pharmacokinetics: Leflunomide is completely absorbed from the gut and has a mean plasma half-life of 19 days. Its active metabolite, A77-1726, has approximately the same half-life and is subject to enterohepatic recirculation. Cholestyramine can enhance leflunomide excretion and increases total clearance by approximately 50%. 3. Indications: Leflunomide is as effective as methotrexate in RA, including inhibition of bony damage. In one study, combined treatment with methotrexate and leflunomide resulted in a 46.2% ACR20 response compared with 19.5% in patients receiving methotrexate alone. 4. Adverse Effects: Diarrhea occurs in approximately 25% of patients given leflunomide, although only about 3–5% of patients discontinue the drug because of this side effect. Elevation in liver enzymes can occur. Both effects can be reduced by decreasing the dose of leflunomide. Other adverse effects associated with leflunomide are mild alopecia, weight gain, and increased blood pressure. Leukopenia and thrombocytopenia occur rarely. This drug is contraindicated in pregnancy.

METHOTREXATE Methotrexate, a synthetic nonbiologic antimetabolite, is the first-line DMARD for treating RA and is used in 50–70% of patients. It is active in this condition at much lower doses than those needed in cancer chemotherapy (see Chapter 54). 1. Mechanism of Action: Methotrexate’s principal mechanism of action at the low doses used in the rheumatic diseases probably relates to inhibition of amino-imidazolecarboxamide ribonucleotide (AICAR) transformylase and thymidylate synthetase. AICAR, which accumulates intracellularly, competitively inhibits AMP deaminase, leading to an accumulation of AMP. The AMP is released and converted extracellularly to adenosine, which is a potent inhibitor of inflammation. As a result, the inflammatory functions of neutrophils, macrophages, dendritic cells, and lymphocytes are suppressed. Methotrexate has secondary effects on polymorphonuclear chemotaxis. There is some effect on dihydrofolate reductase and this affects lymphocyte and macrophage function, but this is not its principal mechanism of action. Methotrexate has direct inhibitory effects on proliferation and stimulates apoptosis in immune-inflammatory cells. Additionally, it inhibits proinflammatory cytokines linked to rheumatoid synovitis. 2. Pharmacokinetics: The drug is approximately 70% absorbed after oral administration (see Chapter 54). It is metabolized to a less active hydroxylated product. Both the parent compound and the metabolite are polyglutamated within cells where they stay for prolonged periods. Methotrexate’s serum half-life is usually only 6–9 hours. Hydroxychloroquine can reduce the clearance or increase the tubular reabsorption of methotrexate. Methotrexate is excreted principally in the urine, but up to 30% may be excreted in bile. 3. Indications: Although the most common methotrexate dosing regimen for the treatment of RA is 15–25 mg weekly, there is an


increased effect up to 30–35 mg weekly. The drug decreases the rate of appearance of new erosions. Evidence supports its use in juvenile chronic arthritis, and it has been used in psoriasis, PA, AS, polymyositis, dermatomyositis, Wegener’s granulomatosis, giant cell arteritis, SLE, and vasculitis. 4. Adverse Effects: Nausea and mucosal ulcers are the most common toxicities. Additionally, many other side effects such as leukopenia, anemia, stomatitis, GI ulcerations, and alopecia are probably the result of inhibiting cellular proliferation. Progressive doserelated hepatotoxicity in the form of enzyme elevation occurs frequently, but cirrhosis is rare (<1%). Liver toxicity is not related to serum methotrexate concentrations. A rare hypersensitivity-like lung reaction with acute shortness of breath has been documented, as have pseudo-lymphomatous reactions. The incidence of GI and liver function test abnormalities can be reduced by the use of leucovorin 24 hours after each weekly dose or by the use of folic acid, although this may decrease the efficacy of the methotrexate by about 10%. This drug is contraindicated in pregnancy.

MYCOPHENOLATE MOFETIL 1. Mechanism of Action: Mycophenolate mofetil (MMF), a semisynthetic DMARD, is converted to mycophenolic acid, the active form of the drug. The active product inhibits inosine monophosphate dehydrogenase, leading to suppression of T- and B-lymphocyte proliferation. Downstream, it interferes with leukocyte adhesion to endothelial cells through inhibition of E-selectin, P-selectin, and intercellular adhesion molecule 1. MMF’s pharmacokinetics and toxicities are discussed in Chapter 55. 2. Indications: MMF is effective for the treatment of renal disease due to SLE and may be useful in vasculitis and Wegener’s granulomatosis. Although MMF is occasionally used at a dosage of 2 g/d to treat RA, there are no well-controlled data regarding its efficacy in this disease. 3. Adverse Effects: MMF is associated with nausea, dyspepsia, and abdominal pain. Like azathioprine, it can cause hepatotoxicity. MMF can also cause leukopenia, thrombocytopenia, and anemia. MMF is associated with an increased incidence of infections. It is only rarely associated with malignancy.

RITUXIMAB 1. Mechanism of Action: Rituximab is a chimeric monoclonal antibody biologic agent that targets CD20 B lymphocytes (see Chapter 55). Depletion of these cells takes place through cell-mediated and complement-dependent cytotoxicity and stimulation of cell apoptosis. Depletion of B lymphocytes reduces inflammation by decreasing the presentation of antigens to T lymphocytes and inhibiting the secretion of proinflammatory cytokines. Rituximab rapidly depletes peripheral B cells, although this depletion correlates neither with efficacy nor with toxicity. 2. Pharmacokinetics: Rituximab is given as two intravenous infusions of 1000 mg, separated by 2 weeks. It may be repeated every 6– 9 months, as needed. Repeated courses remain effective. Pretreatment with acetaminophen, an antihistamine, and intravenous glucocorticoids (usually 100 mg of methylprednisolone) given 30 minutes prior to infusion decreases the incidence and severity of infusion reactions. 3. Indications: Rituximab is indicated for the treatment of moderately to severely active RA in combination with methotrexate in patients with an inadequate response to one or more TNF-α antagonists. Rituximab in combination with glucocorticoids is also approved for the treatment of adult patients with Wegener’s granulomatosis (also known as granulomatosis with polyangiitis) and microscopic polyangiitis and is used in other forms of vasculitis as well (see Chapter 54 for its use in lymphomas and leukemias). 4. Adverse Effects: About 30% of patients develop rash with the first 1000 mg treatment; this incidence decreases to about 10% with the second infusion and progressively decreases with each course of therapy thereafter. These rashes do not usually require discontinuation of therapy, although an urticarial or anaphylactoid reaction precludes further therapy. Immunoglobulins (particularly IgG and IgM) may decrease with repeated courses of therapy and infections can occur, although they do not seem directly associated with the decreases in immunoglobulins. Serious, and sometimes fatal, bacterial, fungal, and viral infections are reported for up to one year of the last dose of rituximab, and patients with severe and active infections should not receive rituximab. Rituximab is associated with reactivation of hepatitis B virus (HBV) infection, which requires monitoring before and several months after the initiation of the treatment. Rituximab has not been associated with either activation of tuberculosis or the occurrence of lymphomas or other tumors (see Chapter 55). Fatal mucocutaneous reactions have been reported in patients receiving rituximab. Different cytopenias can occur, which require complete blood cell monitoring every 2–4 months in RA patients. Other adverse effects, such as cardiovascular events, are rare.

SULFASALAZINE 1. Mechanism of Action: Sulfasalazine, a synthetic nonbiologic DMARD, is metabolized to sulfapyridine and 5-aminosalicylic acid.


The sulfapyridine is probably the active moiety when treating RA (unlike inflammatory bowel disease, see Chapter 62). Some authorities believe that the parent compound, sulfasalazine, also has an effect. Suppression of T-cell responses to concanavalin and inhibition of in vitro B-cell proliferation are documented. In vitro, sulfasalazine or its metabolites inhibit the release of inflammatory cytokines produced by monocytes or macrophages, eg, IL-1, -6, and -12, and TNF-α. 2. Pharmacokinetics: Only 10–20% of orally administered sulfasalazine is absorbed, although a fraction undergoes enterohepatic recirculation into the bowel where it is reduced by intestinal bacteria to liberate sulfapyridine and 5-aminosalicylic acid (see Figure 62–8). Sulfapyridine is well absorbed while 5-aminosalicylic acid remains unabsorbed. Some sulfasalazine is excreted unchanged in the urine whereas sulfapyridine is excreted after hepatic acetylation and hydroxylation. Sulfasalazine’s half-life is 6–17 hours. 3. Indications: Sulfasalazine is effective in RA and reduces radiologic disease progression. It has also been used in juvenile chronic arthritis, PA, inflammatory bowel disease, AS, and spondyloarthropathy-associated uveitis. The usual regimen is 2–3 g/d. 4. Adverse Effects: Approximately 30% of patients using sulfasalazine discontinue the drug because of toxicity. Common adverse effects include nausea, vomiting, headache, and rash. Hemolytic anemia and methemoglobinemia also occur, but rarely. Neutropenia occurs in 1–5% of patients, while thrombocytopenia is very rare. Pulmonary toxicity and positive double-stranded DNA (dsDNA) are occasionally seen, but drug-induced lupus is rare. Reversible infertility occurs in men, but sulfasalazine does not affect fertility in women. The drug does not appear to be teratogenic.

TOCILIZUMAB 1. Mechanism of Action: Tocilizumab, a newer biologic humanized antibody, binds to soluble and membrane-bound IL-6 receptors, and inhibits the IL-6-mediated signaling via these receptors. IL-6 is a proinflammatory cytokine produced by different cell types including T cells, B cells, monocytes, fibroblasts, and synovial and endothelial cells. IL-6 is involved in a variety of physiologic processes such as T-cell activation, hepatic acute-phase protein synthesis, and stimulation of the inflammatory processes involved in diseases such as RA. 2. Pharmacokinetics: The half-life of tocilizumab is dose-dependent, approximately 11 days for the 4 mg/kg dose and 13 days for the 8 mg/kg dose. IL-6 can suppress several CYP450 isoenzymes; thus, inhibiting IL-6 may restore CYP450 activities to higher levels. This may be clinically relevant for drugs that are CYP450 substrates and have a narrow therapeutic window (eg, cyclosporine or warfarin), and dosage adjustment of these medications may be needed. Tocilizumab can be used in combination with nonbiologic DMARDs or as monotherapy. In the USA the recommended starting dose for RA is 4 mg/kg intravenously every 4 weeks followed by an increase to 8 mg/kg (not exceeding 800 mg/infusion) dependent on clinical response. In Europe, the starting dose of tocilizumab is 8 mg/kg up to 800 mg. Tocilizumab dosage in SJIA or PJIA follows an algorithm that accounts for body weight. Additionally, dosage modifications are recommended on the basis of certain laboratory changes such as elevated liver enzymes, neutropenia, and thrombocytopenia. 3. Indications: Tocilizumab is indicated for adult patients with moderately to severely active RA who have had an inadequate response to one or more DMARDs. It is also indicated in patients who are older than 2 years with active SJIA or active PJIA. A recent study showed that it is slightly more effective than adalimumab. 4. Adverse Effects: Serious infections including tuberculosis, fungal, viral, and other opportunistic infections have occurred. Screening for tuberculosis should be done prior to beginning tocilizumab. The most common adverse reactions are upper respiratory tract infections, headache, hypertension, and elevated liver enzymes. Neutropenia and reduction in platelet counts occur occasionally, and lipids (eg, cholesterol, triglycerides, LDL, and HDL) should be monitored. GI perforation has been reported when using tocilizumab in patients with diverticulitis and in those using corticosteroids, although it is not clear that this adverse effect is more common than with TNF-α-blocking agents. Demyelinating disorders including multiple sclerosis are rarely associated with tocilizumab use. Fewer than 1% of the patients taking tocilizumab develop anaphylactic reaction. Anti-tocilizumab antibodies develop in 2% of the patients, and these can be associated with hypersensitivity reactions requiring discontinuation.

TNF-α-BLOCKING AGENTS Cytokines play a central role in the immune response (see Chapter 55) and in RA. Although a wide range of cytokines are expressed in the joints of RA patients, TNF-α appears to be particularly important in the inflammatory process. TNF-α affects cellular function via activation of specific membrane-bound TNF receptors (TNFR1 , TNFR2 ). Five biologic DMARDs interfering with TNF-α have been approved for the treatment of RA and other rheumatic diseases (Figure 36–4). These drugs have many adverse effects in common; these effects are discussed at the end of this section.


FIGURE 36–4 Structures of TNF-ι antagonists used in rheumatoid arthritis. CH, constant heavy chain; CL, constant light chain; Fc, complex immunoglobulin region; VH, variable heavy chain; VL, variable light chain. Red regions, human derived; blue regions, mouse derived; green regions, polyethylene glycol (PEG).


Adalimumab 1. Mechanism of Action: Adalimumab is a fully human IgG1 anti-TNF monoclonal antibody. This compound complexes with soluble TNF-α and prevents its interaction with p55 and p75 cell surface receptors. This results in down-regulation of macrophage and T-cell function. 2. Pharmacokinetics: Adalimumab is given subcutaneously and has a half-life of 10–20 days. Its clearance is decreased by more than 40% in the presence of methotrexate, and the formation of human anti-monoclonal antibody is decreased when methotrexate is given at the same time. The usual dose in RA is 40 mg every other week, but dosing is frequently increased to 40 mg weekly. In psoriasis, 80 mg is given at week 0, 40 mg at week 1, and then 40 mg every other week thereafter. The initial dose in inflammatory bowel disease is higher; patients receive 160 mg at week 0, 80 mg 2 weeks later, followed by a 40 mg maintenance dose every other week. Patients with ulcerative colitis should continue maintenance treatment beyond 8 weeks if they show evidence of remission by that time. Adalimumab dose depends on the body weight in patients with JIA; 20 mg every other week for patients weighing 15–30 kg, and 40 mg every other week in patients weighing 30 kg or more. 3. Indications: The compound is approved for RA, AS, PA, JIA, plaque psoriasis, Crohn’s disease, and ulcerative colitis. It decreases the rate of formation of new erosions. It is effective both as monotherapy and in combination with methotrexate and other nonbiologic DMARDs. Based only on case reports and case series, adalimumab has also been found to be effective in the treatment of Behçet’s disease, sarcoidosis, and notably, noninfectious uveitis.

Certolizumab 1. Mechanism of Action: Certolizumab is a recombinant, humanized antibody Fab fragment conjugated to a polyethylene glycol (PEG) with specificity for human TNF-α. Certolizumab neutralizes membrane-bound and soluble TNF-α in a dose-dependent manner. Additionally, certolizumab does not contain an Fc region, found on a complete antibody, and does not fix complement or cause antibody-dependent cell-mediated cytotoxicity in vitro. 2. Pharmacokinetics: Certolizumab is given subcutaneously and has a half-life of 14 days. Methotrexate decreases the appearance of anti-certolizumab antibodies. The usual dose for RA is 400 mg initially and at weeks 2 and 4, followed by 200 mg every other week, or 400 mg every 4 weeks. 3. Indications: Certolizumab is indicated for the treatment of adults with moderately to severely active RA. It can be used as monotherapy or in combination with nonbiologic DMARDs. Additionally, certolizumab is approved in adult patients with Crohn’s disease, active PA and active AS.

Etanercept 1. Mechanism of Action: Etanercept is a recombinant fusion protein consisting of two soluble TNF p75 receptor moieties linked to the Fc portion of human IgG1 (Figure 36–4); it binds TNF-α molecules and also inhibits lymphotoxin-α. 2. Pharmacokinetics: Etanercept is given subcutaneously as 25 mg twice weekly or 50 mg weekly. In psoriasis, 50 mg is given twice weekly for 12 weeks and then is followed by 50 mg weekly. The drug is slowly absorbed, with peak concentration 72 hours after drug administration. Etanercept has a mean serum elimination half-life of 4.5 days. A recent study demonstrated a reduction of radiographic progression with the use of 50 mg of etanercept weekly. 3. Indications: Etanercept is approved for the treatment of RA, juvenile chronic arthritis, psoriasis, PA, and AS. It can be used as monotherapy, although over 70% of patients taking etanercept are also using methotrexate. Etanercept decreases the rate of formation of new erosions relative to methotrexate alone. It is also being used in other rheumatic syndromes such as scleroderma, granulomatosis with polyangiitis (Wegener’s granulomatosis), giant cell arteritis, Behçet’s disease, uveitis, and sarcoidosis.

Golimumab 1. Mechanism of Action: Golimumab is a human monoclonal antibody with a high affinity for soluble and membrane-bound TNF-α. Golimumab effectively neutralizes the inflammatory effects produced by TNF-α seen in diseases such as RA. 2. Pharmacokinetics: Golimumab is administered subcutaneously and has a half-life of approximately 14 days. Concomitant use with methotrexate increases golimumab serum levels and decreases anti-golimumab antibodies. The recommended dose for the treatment of RA, PA, and AS is 50 mg given every 4 weeks. A higher dose of golimumab is used for the treatment of ulcerative colitis as follows: 200 mg initially at week 0 followed by 100 mg at week 2 and every 4 weeks thereafter. 3. Indications: Golimumab with methotrexate is indicated for the treatment of moderately to severely active RA in adult patients. It is also indicated for the treatment of PA and AS and moderate to severe ulcerative colitis.


Infliximab 1. Mechanism of Action: Infliximab (Figure 36–4) is a chimeric (25% mouse, 75% human) IgG1 monoclonal antibody that binds with high affinity to soluble and possibly membrane-bound TNF-α. Its mechanism of action probably is the same as that of adalimumab. 2. Pharmacokinetics: Infliximab is given as an intravenous infusion with “induction” at 0, 2, and 6 weeks and maintenance every 8 weeks thereafter. Dosing is 3–10 mg/kg, and the usual dose is 3–5 mg/kg every 8 weeks. There is a relationship between serum concentration and effect, although individual clearances vary markedly. The terminal half-life is 9–12 days without accumulation after repeated dosing at the recommended interval of 8 weeks. After intermittent therapy, infliximab elicits human antichimeric antibodies in up to 62% of patients. Concurrent therapy with methotrexate markedly decreases the prevalence of human antichimeric antibodies. 3. Indications: Infliximab is approved for use in RA, AS, PA, Crohn’s disease, ulcerative colitis, pediatric inflammatory bowel disease, and psoriasis. It is being used off-label in other diseases, including granulomatosis with polyangiitis (Wegener’s granulomatosis), giant cell arteritis, Behçet’s disease, uveitis, and sarcoidosis. In RA, infliximab plus methotrexate decreases the rate of formation of new erosions. Although it is recommended that methotrexate be used in conjunction with infliximab, a number of other nonbiologic DMARDs, including antimalarials, azathioprine, leflunomide, and cyclosporine, can be used as background therapy for this drug. Infliximab is also used as monotherapy.

Adverse Effects of TNF-α-Blocking Agents TNF-α-blocking agents have multiple adverse effects in common. The risk of bacterial infections and macrophage-dependent infection (including tuberculosis, fungal, and other opportunistic infections) is increased, although it remains very low. Activation of latent tuberculosis is lower with etanercept than with other TNF-α-blocking agents. Nevertheless, all patients should be screened for latent or active tuberculosis before starting TNF-α-blocking agents. The use of TNF-α-blocking agents is also associated with increased risk of HBV reactivation and screening for HBV is important before starting the treatment. TNF-α-blocking agents increase the risk of skin cancers—including melanoma—which necessitates periodic skin examination, especially in high-risk patients. On the other hand, there is no clear evidence of increased risk of solid malignancies or lymphomas with TNF-α-blocking agents, and their incidence may not be different compared with other DMARDs or active RA itself. A low incidence of newly formed dsDNA antibodies and antinuclear antibodies (ANAs) has been documented when using TNF-αblocking agents, but clinical lupus is extremely rare and the presence of ANA and dsDNA antibodies per se does not contraindicate the use of TNF-α-blocking agents. In patients with borderline or overt heart failure (HF), TNF-α-blocking agents can exacerbate HF. TNFα-blocking agents can induce the immune system to develop anti-drug antibodies in about 17% of cases. These antibodies may interfere with drug efficacy and correlate with infusion site reactions. Injection site reactions occur in 20–40% of patients, although they rarely result in discontinuation of therapy. Cases of alopecia areata, hypertrichosis, and erosive lichen planus have been reported. Cutaneous pseudo lymphomas are reported rarely with TNF-α-blocking agents, especially infliximab. TNF-α-blocking agents may increase the risk of gastrointestinal ulcers and large bowel perforation including diverticular and appendiceal perforation. Nonspecific interstitial pneumonia, psoriasis, and sarcoidosis-like syndrome are among the rare reported toxicities associated with TNF-α blockers. Rare cases of leukopenia, neutropenia, thrombocytopenia, and pancytopenia have been reported. The precipitating drug should be discontinued in such cases.

TOFACITINIB 1. Mechanism of Action: Tofacitinib is a synthetic small molecule that selectively inhibits all members of the Janus kinase (JAK, see Chapter 2) family to varying degrees. At therapeutic doses, tofacitinib exerts its effect mainly by inhibiting JAK3, and to a lesser extent JAK1, hence interrupting the JAK-STAT signaling pathway. This pathway plays a major role in the pathogenesis of autoimmune diseases including RA. The JAK3/JAK1 complex is responsible for signal transduction from the common γ-chain receptor (IL-2RG) for IL-2, -4, -7, -9, -15, and -21, which subsequently influences transcription of several genes that are crucial for the differentiation, proliferation, and function of NK cells and T and B lymphocytes. In addition, JAK1 (in combination with other JAKs) controls signal transduction from IL-6 and interferon receptors. RA patients receiving tofacitinib rapidly reduce the C-reactive protein. 2. Pharmacokinetics: Tofacitinib is an oral, targeted DMARD. The recommended dose in the treatment of RA is 5 mg twice daily; there is a clear trend to increased response (and increased toxicity) at double this dose. Tofacitinib has an absolute oral bioavailability of 74%, high-fat meals do not affect the AUC, and the elimination half-life is about 3 hours. Metabolism (of 70%) occurs in the liver, mainly by CYP3A4 and to a lesser extent by CYP2C19. The remaining 30% is excreted unchanged by the kidneys. Patients taking CYP enzyme inhibitors and those with moderate hepatic or renal impairment require dose reduction to 5 mg once daily. It should not be given to patients with severe hepatic disease. 3. Indications: Tofacitinib was originally developed to prevent solid organ allograft rejection. It has also been tested for the treatment


of inflammatory bowel disease, spondyloarthritis, psoriasis, and dry eyes. To date, tofacitinib is approved in the USA for the treatment of adult patients with moderately to severely active RA who have failed or are intolerant to methotrexate. It is not approved in Europe for this indication. It can be used as a monotherapy or in combination with other nonbiologic DMARDs, including methotrexate. 4. Adverse Effects: As with biologic DMARDs, tofacitinib slightly increases the risk of infection, and it should not be used with potent immunosuppressants or biologic DMARDs due to added immunosuppressive effects. Upper respiratory tract infection and urinary tract infection represent the most common infections. More serious infections are also reported, including pneumonia, cellulitis, esophageal candidiasis, and other opportunistic infections. All patients should be screened for latent or active tuberculosis before the initiation of treatment. Lymphoma and other malignancies such as lung and breast cancer have been reported in patients taking tofacitinib, although some studies discuss the potential use of JAK inhibitors to treat certain lymphomas. Dose-dependent increases in the levels of low-density lipoprotein (LDL), high-density lipoprotein (HDL), and total cholesterol have been found in patients receiving tofacitinib, often beginning about 6 weeks after starting treatment; therefore, lipid levels should be monitored. Although tofacitinib causes a dose-dependent increase in CD19 B cells and CD4 T cells plus a reduction in CD16/CD56 NK cells, the clinical significance of these changes remains unclear. Drug-related neutropenia and anemia occur, requiring drug discontinuation. Headache, diarrhea, elevation of liver enzymes, and gastrointestinal perforation are among the other reported effects of tofacitinib.

INTERLEUKIN-1 INHIBITORS IL-1α plays a major role in the pathogenesis of several inflammatory and autoimmune diseases including RA. IL-1β and IL-1 receptor antagonist (IL-1RA) are other members of the IL-1 family. All three bind to IL-1 receptors in the same manner. However, IL-1RA does not initiate the intracellular signaling pathway and thus acts as a competitive inhibitor of the proinflammatory IL-1α and IL-1β.

Anakinra 1. Mechanism of Action: Anakinra is the oldest drug in this family but is now rarely used for RA. Anakinra is a recombinant IL-1RA; it blocks the effect of IL-1α and IL-1β on IL-1 receptors, hence decreasing the immune response in inflammatory diseases. 2. Pharmacokinetics: Anakinra is administered subcutaneously and reaches a maximum plasma concentration after 3–7 hours. The absolute bioavailability of anakinra is 95%, and it has a 4- to 6-hour terminal half-life. The recommended dose in the treatment of RA is 100 mg daily. The dose of anakinra depends on the body weight in the treatment of cryopyrin-associated periodic syndrome (CAPS), starting with 1–2 mg/kg/d to a maximum of 8 mg/kg/d. Reduction in the frequency of administering anakinra to every other day is recommended in patients with renal insufficiency. 3. Indications: Anakinra is approved for the treatment of moderately to severely active RA in adult patients, but it is not very effective and is rarely used for this indication. However, anakinra is the drug of choice for CAPS, particularly the neonatal-onset multisystem inflammatory disease (NOMID) subtype. Anakinra is effective in gout (see below) and is used for other diseases including Behçet’s disease and adult onset JIA. Its use for giant cell arteritis is controversial.

Canakinumab 1. Mechanism of Action: Canakinumab is a human IgG1 /κ monoclonal antibody against IL-1β. It forms a complex with IL-1β, preventing its binding to IL-1 receptors. 2. Pharmacokinetics: Canakinumab is given as subcutaneous injections. It reaches peak serum concentrations 7 days after a single subcutaneous injection. Canakinumab has an absolute bioavailability of 66% and a 26-day mean terminal half-life. The recommended dose for patients with SJIA who weigh more than 7.5 kg is 4 mg/kg every 4 weeks. There is a weight-adjusted algorithm for treating CAPS. 3. Indications: Canakinumab is indicated for active SJIA in children 2 years or older. It is also used to treat CAPS, particularly the familial cold autoinflammatory syndrome and Muckle-Wells syndrome subtypes for adults and children 4 years or older. Canakinumab is also used to treat gout (see below).

Rilonacept 1. Mechanism of Action: Rilonacept is the ligand-binding domain of the IL-1 receptor. It binds mainly to IL-1β and binds with lower affinity to IL-1α and IL-1RA. Rilonacept neutralizes IL-1β and prevents its attachment to IL-1 receptors. 2. Pharmacokinetics: The subcutaneous dose of rilonacept for CAPS is age-dependent. In patients 12–17 years of age, 4.4 mg/kg (maximum of 320 mg) is the loading dose, with a maintenance dose of 2.2 mg/kg (maximum of 160 mg) weekly. Those 18 years and older receive 320 mg as a loading dose and 160 mg weekly thereafter. The steady-state plasma concentration is reached after 6


weeks. 3. Indications: Rilonacept is approved to treat CAPS subtypes: familial cold autoinflammatory syndrome and Muckle-Wells syndrome in patients 12 years or older. Rilonacept is also used to treat gout (see below).

Adverse Effects of Interleukin-1 Inhibitors The most common adverse effects are injection site reactions (up to 40%) and upper respiratory tract infections. Serious infections occur rarely in patients given IL-1 inhibitors. Headache, abdominal pain, nausea, diarrhea, arthralgia, and flu-like illness have all been reported, as well as hypersensitivity reactions. Patients taking IL-1 inhibitors may experience transient neutropenia, which requires regular monitoring of neutrophil counts.

BELIMUMAB Belimumab is an antibody that specifically inhibits B-lymphocyte stimulator (BLyS). It is administered as an intravenous infusion. The recommended dose is 10 mg/kg at weeks 0, 2, 4, and every 4 weeks thereafter. Belimumab has a distribution half-life of 1.75 days and a terminal half-life of 19.4 days. Belimumab is approved only for the treatment of adult patients with active, seropositive SLE who are receiving standard treatment. The drug was approved after a protracted series of clinical trials, and its place in the SLE armamentarium is not clear. Belimumab should not be used in patients with active renal or neurological manifestations of SLE, as there are no data for these conditions. In addition, the efficacy of belimumab has not been tested in combination with other biologic DMARDs or cyclophosphamide. The most common adverse effects of belimumab are nausea, diarrhea, and respiratory tract infection. As with other biologic DMARDs, there is a slight increase in the risk of infection including serious infections. Cases of depression and suicide have been reported in patients receiving belimumab, although these patients may have had neurologic SLE, thus confounding the causal relationship. Infusion reactions including anaphylaxis are among the other adverse effects. A very small percentage of patients develop antibodies toward belimumab; their clinical significance however is not clear.

COMBINATION THERAPY WITH DMARDS In a 1998 survey, approximately half of North American rheumatologists treated moderately aggressive RA with combination therapy, and the use of drug combinations is probably much higher now. Combinations of DMARDs can be designed rationally on the basis of complementary mechanisms of action, nonoverlapping pharmacokinetics, and nonoverlapping toxicities. When added to methotrexate background therapy, cyclosporine, chloroquine, hydroxychloroquine, leflunomide, infliximab, adalimumab, rituximab, and etanercept have all shown improved efficacy. Triple therapy with methotrexate, sulfasalazine, and hydroxychloroquine appears to be as effective as etanercept and methotrexate. In contrast, azathioprine or sulfasalazine plus methotrexate results in no additional therapeutic benefit. Other combinations have occasionally been used. While it might be anticipated that combination therapy could result in more toxicity, this is often not the case. Combination therapy for patients not responding adequately to monotherapy is now the rule in the treatment of RA.

GLUCOCORTICOID DRUGS The general pharmacology of corticosteroids, including mechanism of action, pharmacokinetics, and other applications, is discussed in Chapter 39.

Indications Corticosteroids have been used in 60–70% of RA patients. Their effects are prompt and dramatic, and they are capable of slowing the appearance of new bone erosions. Corticosteroids may be administered for certain serious extra-articular manifestations of RA such as pericarditis or eye involvement or during periods of exacerbation. When prednisone is required for long-term therapy, the dosage should not exceed 7.5 mg daily, and gradual reduction of the dose should be encouraged. Alternate-day corticosteroid therapy is usually unsuccessful in RA. Other rheumatic diseases in which the corticosteroids’ potent anti-inflammatory effects may be useful include vasculitis, SLE, Wegener’s granulomatosis, PA, giant cell arteritis, sarcoidosis, and gout. Intra-articular corticosteroids are often helpful to alleviate painful symptoms and, when successful, are preferable to increasing the dosage of systemic medication. Some of the symptoms of RA, especially morning stiffness and joint pain, follow a circadian rhythm, probably due to an increase in proinflammatory cytokines in the early morning. A recent approach uses delayed-release prednisone for the treatment of early


morning stiffness and pain in RA. The tablet contains an inactive outer layer and a core of the active drug. The outer layer dissolves over 4–6 hours, releasing the prednisone. Taking the drug at 9–10 pm results in a small pulse of prednisone at 2–4 am, decreasing the circadian inflammatory cytokines. At low doses of 3–5 mg prednisone, the adrenal-pituitary axis does not seem to be impacted.

Adverse Effects Prolonged use of corticosteroids leads to serious and disabling toxic effects as described in Chapter 39. Many of these adverse effects occur at doses below 7.5 mg prednisone equivalent daily and many experts believe that even 3–5 mg/d can cause adverse effects in susceptible individuals when this class of drugs is used over prolonged periods.

OTHER ANALGESICS Acetaminophen is one of the most important drugs used in the treatment of mild to moderate pain when an anti-inflammatory effect is not necessary. Phenacetin, a prodrug that is metabolized to acetaminophen, is more toxic and should not be used.

ACETAMINOPHEN Acetaminophen is the active metabolite of phenacetin and is responsible for its analgesic effect. It is a weak COX-1 and COX-2 inhibitor in peripheral tissues and possesses no significant anti-inflammatory effects.

1. Pharmacokinetics: Acetaminophen is administered orally. Peak blood concentrations are usually reached in 30–60 minutes. Acetaminophen is poorly bound to plasma proteins and is partially metabolized by hepatic microsomal enzymes to the inactive sulfate and glucuronide (see Figure 4–5). Less than 5% is excreted unchanged. A minor but highly reactive metabolite (N-acetyl-pbenzoquinone) is important in large doses because it is toxic to both liver and kidney (see Chapter 4). The half-life of acetaminophen is 2–3 hours and is relatively unaffected by renal function. With toxic doses or liver disease, the half-life may be increased twofold or more. 2. Indications: Although said to be equivalent to aspirin as an analgesic and antipyretic agent, acetaminophen lacks anti-inflammatory properties. It does not affect uric acid levels and lacks platelet-inhibiting effects. The drug is useful in mild to moderate pain such as headache, myalgia, postpartum pain, and other circumstances in which aspirin is an effective analgesic. Acetaminophen alone is inadequate therapy for inflammatory conditions such as RA. For mild analgesia, acetaminophen is the preferred drug in patients allergic to aspirin, when salicylates are poorly tolerated. It is preferable to aspirin in patients with hemophilia, in those with a history of peptic ulcer, and in those in whom bronchospasm is precipitated by aspirin. Unlike aspirin, acetaminophen does not antagonize the effects of uricosuric agents. 3. Adverse Effects: In therapeutic doses, a mild reversible increase in hepatic enzymes may occasionally occur. With larger doses, dizziness, excitement, and disorientation may occur. Ingestion of 15 g of acetaminophen may be fatal, death being caused by severe hepatotoxicity with centrilobular necrosis, sometimes associated with acute renal tubular necrosis (see Chapters 4 and 58). 4. Present data indicate that even 4 g acetaminophen is associated with increased liver function test abnormalities. Doses greater than 4 g/d are not usually recommended and a history of alcoholism contraindicates even this dose. Early symptoms of hepatic damage include nausea, vomiting, diarrhea, and abdominal pain. Cases of renal damage without hepatic damage have occurred, even after usual doses of acetaminophen. Therapy for overdose is much less satisfactory than that for aspirin overdose. In addition to supportive therapy, one should provide sulfhydryl groups in the form of acetylcysteine to neutralize the toxic metabolites (see Chapter 58). 5. Hemolytic anemia and methemoglobinemia are very rare adverse events. Interstitial nephritis and papillary necrosis—serious complications of phenacetin—have not occurred, and GI bleeding also has not occurred. Caution is necessary in patients with any type of liver disease. 6. Dosage: Acute pain and fever may be effectively treated with 325–500 mg four times daily and proportionately less for children. Dosing in adults is now recommended not to exceed 4 g/d, in most cases.

KETOROLAC Ketorolac is an NSAID promoted for systemic use mainly as a short-term analgesic (not longer than 1 week), not as an anti-


inflammatory drug (although it has typical NSAID properties). Pharmacokinetics are presented in Table 36–1. The drug is an effective analgesic and has been used successfully to replace morphine in some situations involving mild to moderate postsurgical pain. It is most often given intramuscularly or intravenously, but an oral formulation is available. When used with an opioid, it may decrease the opioid requirement by 25–50%. Toxicities are similar to those of other NSAIDs (see pages 620-621), although renal toxicity is more common with chronic use.

TRAMADOL Tramadol is a centrally acting synthetic analgesic, structurally related to opioids. Since naloxone, an opioid receptor blocker, only inhibits 30% of the analgesic effect of tramadol, the mechanism of action of this drug must involve both nonopioid and opioid receptors. Tramadol does not have significant anti-inflammatory effects. The drug may exert part of its analgesic effect by enhancing 5hydroxytryptamine (5-HT) release and inhibiting the reuptake of norepinephrine and 5-HT (see Chapter 31).

DRUGS USED IN GOUT Gout is a metabolic disease characterized by recurrent episodes of acute arthritis due to deposits of monosodium urate in joints and cartilage. Uric acid renal calculi, tophi, and interstitial nephritis may also occur. Adverse cardiovascular outcomes are becoming more clear as well. Gout is usually associated with a high serum uric acid level (hyperuricemia), a poorly soluble substance that is the major end product of purine metabolism. In most mammals, uricase converts uric acid to the more soluble allantoin; this enzyme is absent in humans. While clinical gouty episodes are associated with hyperuricemia, most individuals with hyperuricemia may never develop a clinical event from urate crystal deposition. The treatment of gout aims to relieve acute gouty attacks and prevent recurrent gouty episodes and urate lithiasis. Therapies for acute gout are based on our current understanding of the pathophysiologic events that occur in this disease (Figure 36–5). Clinical gout is dependent on a macromolecular complex of proteins, called NLRP3, which regulates the activation of IL-1. Urate crystals activate NLRP3, resulting in release of prostaglandins and lysosomal enzymes by synoviocytes. Attracted by these chemotactic mediators, polymorphonuclear leukocytes migrate into the joint space and amplify the ongoing inflammatory process. In the later phases of the attack, increased numbers of mononuclear phagocytes (macrophages) appear, ingest the urate crystals, and release more inflammatory mediators.

FIGURE 36–5 Pathophysiologic events in a gouty joint. Synoviocytes phagocytose urate crystals and then secrete inflammatory mediators, which attract and activate polymorphonuclear leukocytes (PMN) and mononuclear phagocytes (MNP) (macrophages). Drugs active in gout inhibit crystal phagocytosis and polymorphonuclear leukocyte and macrophage release of inflammatory mediators. PG, prostaglandin; IL-1, interleukin-1; LTB4 , leukotriene B4 . Before starting chronic urate-lowering therapy for gout, patients in whom hyperuricemia is associated with gout and urate lithiasis


must be clearly distinguished from individuals with only hyperuricemia. The efficacy of long-term drug treatment in an asymptomatic hyperuricemic person is unproved. Although there are data suggesting a clear relationship between the degree of uric acid elevation and the likelihood of clinical gout, in some individuals, uric acid levels may be elevated up to 2 standard deviations above the mean for a lifetime without adverse consequences. Many different agents have been used for the treatment of acute and chronic gout. However, non-adherence to these drugs is exceedingly common; adherence has been documented to be 18%–26% in younger patients. Providers should be aware of compliance as an important issue.

COLCHICINE Although NSAIDs, corticosteroids, or colchicine are first-line drugs for acute gout, colchicine was the primary treatment for many years. Colchicine is an alkaloid isolated from the autumn crocus, Colchicum autumnale. Its structure is shown in Figure 36–6.


FIGURE 36–6 Colchicine and uricosuric drugs. 1. Pharmacokinetics: Colchicine is absorbed readily after oral administration, reaches peak plasma levels within 2 hours, and is eliminated with a serum half-life of 9 hours. Metabolites are excreted in the intestinal tract and urine. 2. Pharmacodynamics: Colchicine relieves the pain and inflammation of gouty arthritis in 12–24 hours without altering the metabolism or excretion of urates and without other analgesic effects. Colchicine produces its anti-inflammatory effects by binding to the intracellular protein tubulin, thereby preventing its polymerization into microtubules and leading to the inhibition of leukocyte migration and phagocytosis. It also inhibits the formation of leukotriene B4 and IL-1β. Several of colchicine’s adverse effects are produced by its inhibition of tubulin polymerization and cell mitosis. 3. Indications: Colchicine is indicated for gout and is also used between attacks (the “intercritical period”) for prolonged prophylaxis (at low doses). It prevents attacks of acute Mediterranean fever and may have a mild beneficial effect in sarcoid arthritis and in


hepatic cirrhosis. Colchicine is also used to treat and prevent pericarditis, pleurisy, and coronary artery disease, probably due to its anti-inflammatory effect. Although it has been given intravenously, this route is no longer approved by the FDA (2009). 4. Adverse Effects: Colchicine often causes diarrhea and may occasionally cause nausea, vomiting, and abdominal pain. Hepatic necrosis, acute renal failure, disseminated intravascular coagulation, and seizures have also been observed. Colchicine may rarely cause hair loss and bone marrow depression, as well as peripheral neuritis, myopathy, and, in some cases, death. The more severe adverse events have been associated with the intravenous administration of colchicine. 5. Dosage: In prophylaxis (the most common use), the dosage of colchicine is 0.6 mg one to three times daily. For terminating a gouty attack, a regimen of 1.2 mg followed by a single 0.6 mg oral dose was as effective as higher dose regimens and adverse events were less with this lower dose regimen. In 2008, the FDA requested that intravenous preparations containing colchicine be discontinued in the USA because of their potential life-threatening adverse effects. Therefore, intravenous colchicine is no longer available. In 2009, the FDA approved a new oral formulation of colchicine for the treatment of acute gout, allowing Colcrys (a branded colchicine) marketing exclusivity in the USA. Generic colchicine rather than Colcrys is available throughout the rest of the world.

NSAIDS IN GOUT In addition to inhibiting prostaglandin synthase, NSAIDs inhibit urate crystal phagocytosis. Aspirin is not used because it causes renal retention of uric acid at low doses (≤ 2.6 g/d). It is uricosuric at doses greater than 3.6 g/d. Indomethacin is commonly used in the initial treatment of gout as a replacement for colchicine. For acute gout, 50 mg is given three times daily; when a response occurs, the dosage is reduced to 25 mg three times daily for 5–7 days. All other NSAIDs except aspirin, salicylates, and tolmetin have been successfully used to treat acute gouty episodes. Oxaprozin, which lowers serum uric acid, is theoretically a good choice. These agents appear to be as effective and safe as the older drugs.

URICOSURIC AGENTS Probenecid and sulfinpyrazone are uricosuric drugs employed to decrease the body pool of urate in patients with tophaceous gout or in those with increasingly frequent gouty attacks. In a patient who excretes large amounts of uric acid, the uricosuric agents should not be used. Lesinurad (RDEA594) is a promising new uricosuric agent that is currently in phase 3 trials. 1. Chemistry and Pharmacokinetics: Uricosuric drugs are organic acids (Figure 36–6) and, as such, act at the anion transport sites of the renal tubule (see Chapter 15). Probenecid is completely reabsorbed by the renal tubules and is metabolized slowly with a terminal serum half-life of 5–8 hours. Sulfinpyrazone or its active hydroxylated derivative is excreted by the kidneys. Even so, the duration of its effect after oral administration is almost as long as that of probenecid, which is given once or twice daily. 2. Pharmacodynamics: Uricosuric drugs—probenecid, sulfinpyrazone, fenofibrate, and losartan—inhibit active transport sites for reabsorption and secretion in the proximal renal tubule so that net reabsorption of uric acid in the proximal tubule is decreased. Because aspirin in doses of less than 2.6 g daily causes net retention of uric acid by inhibiting the secretory transporter, it should not be used for analgesia in patients with gout. The secretion of other weak acids (eg, penicillin) is also reduced by uricosuric agents. 3. As the urinary excretion of uric acid increases, the size of the urate pool decreases, although the plasma concentration may not be greatly reduced. In patients who respond favorably, tophaceous deposits of urate are reabsorbed, with relief of arthritis and remineralization of bone. With the ensuing increase in uric acid excretion, a predisposition to the formation of renal stones is augmented rather than decreased; therefore, the urine volume should be maintained at a high level, and at least early in treatment, the urine pH should be kept above 6.0 by the administration of alkali. 4. Indications: Uricosuric therapy should be initiated in gouty patients with underexcretion of uric acid when allopurinol or febuxostat is contraindicated or when tophi are present. Therapy should not be started until 2–3 weeks after an acute attack. 5. Adverse Effects: Both of these organic acids cause equivalent GI irritation, but sulfinpyrazone is more active in this regard. A rash may appear after the use of either compound. Nephrotic syndrome has occurred after the use of probenecid. Both sulfinpyrazone and probenecid may rarely cause aplastic anemia. 6. Contraindications and Cautions: It is essential to maintain a large urine volume to minimize the possibility of stone formation. 7. Dosage: Probenecid is usually started at a dosage of 0.5 g orally daily in divided doses, progressing to 1 g daily after 1 week. Sulfinpyrazone is started at a dosage of 200 mg orally daily, progressing to 400–800 mg daily. It should be given in divided doses with food to reduce adverse GI effects.

ALLOPURINOL The preferred and standard-of-care therapy for gout during the period between acute episodes is allopurinol, which reduces total uric


acid body burden by inhibiting xanthine oxidase. 1. Chemistry and Pharmacokinetics: The structure of allopurinol, an isomer of hypoxanthine, is shown in Figure 36–7. Allopurinol is approximately 80% absorbed after oral administration and has a terminal serum half-life of 1–2 hours. Like uric acid, allopurinol is metabolized by xanthine oxidase, but the resulting compound, alloxanthine, retains the capacity to inhibit xanthine oxidase and has a long enough duration of action so that allopurinol is given only once a day.

FIGURE 36–7 Inhibition of uric acid synthesis by allopurinol occurs because allopurinol and alloxanthine inhibit xanthine oxidase. (Reproduced, with permission, from Meyers FH, Jawetz E, Goldfien A: Review of Medical Pharmacology, 7th ed. McGraw-Hill, 1980. Copyright © The McGraw-Hill Companies, Inc.) 2. Pharmacodynamics: Dietary purines are not an important source of uric acid. Quantitatively important amounts of purine are formed from amino acids, formate, and carbon dioxide in the body. Those purine ribonucleotides not incorporated into nucleic acids and derived from nucleic acid degradation are converted to xanthine or hypoxanthine and oxidized to uric acid (Figure 36–7). Allopurinol inhibits this last step, resulting in a fall in the plasma urate level and a decrease in the overall urate burden. The more soluble xanthine and hypoxanthine are increased. 3. Indications: Allopurinol is often the first-line agent for the treatment of chronic gout in the period between attacks and it tends to prolong the intercritical period. As with uricosuric agents, the therapy is begun with the expectation that it will be continued for years if not for life. When initiating allopurinol, colchicine or NSAID should be used until steady-state serum uric acid is normalized or decreased to less than 6 mg/dL and they should be continued for 6 months or longer. Thereafter, colchicine or the NSAID can be cautiously stopped while continuing allopurinol therapy. 4. Adverse Effects: In addition to precipitating gout (the reason to use concomitant colchicine or NSAID), GI intolerance (including nausea, vomiting, and diarrhea), peripheral neuritis and necrotizing vasculitis, bone marrow suppression, and aplastic anemia may rarely occur. Hepatic toxicity and interstitial nephritis have been reported. An allergic skin reaction characterized by pruritic maculopapular lesions occurs in 3% of patients. Isolated cases of exfoliative dermatitis have been reported. In very rare cases, allopurinol has become bound to the lens, resulting in cataracts. 5. Interactions and Cautions: When chemotherapeutic purines (eg, azathioprine) are given concomitantly with allopurinol, their dosage must be reduced by about 75%. Allopurinol may also increase the effect of cyclophosphamide. Allopurinol inhibits the metabolism of probenecid and oral anticoagulants and may increase hepatic iron concentration. Safety in children and during pregnancy has not been established. 6. Dosage: The initial dosage of allopurinol is 50–100 mg/d. It should be titrated upward until serum uric acid is below 6 mg/dL; this level is commonly achieved at 300–400 mg/d but is not restricted to this dose; doses as high as 800 mg/d may be needed.


As noted above, colchicine or an NSAID should be given during the first months of allopurinol therapy to prevent the gouty arthritis episodes that sometimes occur.

FEBUXOSTAT Febuxostat is a non-purine xanthine oxidase inhibitor that was approved by the FDA in 2009. 1. Pharmacokinetics: Febuxostat is more than 80% absorbed following oral administration. With maximum concentration achieved in approximately 1 hour and a half-life of 4–18 hours, once-daily dosing is effective. Febuxostat is extensively metabolized in the liver. All of the drug and its inactive metabolites appear in the urine, although less than 5% appears as unchanged drug. 2. Pharmacodynamics: Febuxostat is a potent and selective inhibitor of xanthine oxidase, thereby reducing the formation of xanthine and uric acid without affecting other enzymes in the purine or pyrimidine metabolic pathway. In clinical trials, Febuxostat at daily dosing of 80 mg or 120 mg was more effective in lowering serum urate levels than allopurinol at a standard 300 mg daily dose. The urate-lowering effect was comparable regardless of the pathogenic cause of hyperuricemia—overproduction or underexcretion. 3. Indications: Febuxostat is approved at doses of 40 or 80 mg for the treatment of chronic hyperuricemia in gout patients. Although it appeared to be more effective then allopurinol as urate-lowering therapy, the allopurinol dosing was limited to 300 mg/d, thus not reflecting the actual dosing regimens used in clinical practice. At this time, the dose equivalence of allopurinol and febuxostat is unknown. 4. Adverse Effects: As with allopurinol, prophylactic treatment with colchicine or NSAIDs should be started at the beginning of therapy to avoid gout flares. The most frequent treatment-related adverse events are liver function abnormalities, diarrhea, headache, and nausea. Febuxostat is well tolerated in patients with a history of allopurinol intolerance. There does not appear to be an increased risk of cardiovascular events. 5. Dosage: The recommended starting dose of febuxostat is 40 mg daily. Because there was concern for cardiovascular events in the original phase 3 trials, the FDA approved only 40 mg and 80 mg dosing. No dose adjustment is necessary for patients with renal impairment since it is highly metabolized into an inactive metabolite by the liver.

PEGLOTICASE Pegloticase is the newest urate-lowering therapy to be approved for the treatment of refractory chronic gout. 1. Chemistry: Pegloticase is a recombinant mammalian uricase that is covalently attached to methoxy polyethylene glycol (mPEG) to prolong the circulating half-life and diminish immunogenic response. 2. Pharmacokinetics and Dosage: The recommended dose for pegloticase is 8 mg every 2 weeks administered as an intravenous infusion. It is a rapidly acting drug, achieving a peak decline in uric acid level within 24–72 hours. The serum half-life ranges from 6 to 14 days. Several studies have shown earlier clearance of PEG-uricase (mean of 11 days) due to antibody response when compared to PEG-uricase antibody-negative subjects (mean of 16.1 days). 3. Pharmacodynamics: Urate oxidase enzyme, absent in humans and some higher primates, converts uric acid to allantoin. This product is highly soluble and can be easily eliminated by the kidney. Pegloticase has been shown to maintain low urate levels for up to 21 days after a single dose at doses of 4–12 mg, allowing for IV dosing every 2 weeks. Pegloticase should not be used for asymptomatic hyperuricemia. 4. Adverse Effects: Gout flare can occur during treatment with pegloticase, especially during the first 3–6 months of treatment, requiring prophylaxis with NSAIDs or colchicine. Large numbers of patients show immune responses to pegloticase. The presence of antipegloticase antibodies is associated with shortened circulating half-life, loss of response leading to a rise in plasma urate levels, and a higher rate of infusion reactions and anaphylaxis. Anaphylaxis occurs in more than 6–15% of patients receiving pegloticase. Monitoring of plasma uric acid level, with rising level as an indicator of antibody production, allows for safer administration and monitoring of efficacy. In addition, other oral urate-lowering agents should be avoided in order not to mask the loss of pegloticase efficacy. Nephrolithiasis, arthralgia, muscle spasm, headache, anemia, and nausea may occur. Other less frequent side effects noted include upper respiratory tract infection, peripheral edema, urinary tract infection, and diarrhea. There is some concern for hemolytic anemia in patients with glucose-6-phosphate dehydrogenase deficiency because of the formation of hydrogen peroxide by uricase; therefore, pegloticase should be avoided in these patients.

GLUCOCORTICOIDS Corticosteroids are sometimes used in the treatment of severe symptomatic gout, by intra-articular, systemic, or subcutaneous routes, depending on the degree of pain and inflammation.


The most commonly used oral corticosteroid is prednisone. The recommended oral dose is 30–50 mg/d for 1–2 days, tapered over 7– 10 days. Intra-articular injection of 10 mg (small joints), 30 mg (wrist, ankle, elbow), and 40 mg (knee) of triamcinolone acetonide can be given if the patient is unable to take oral medications.

INTERLEUKIN-1 INHIBITORS Drugs targeting the IL-1 pathway, such as anakinra, canakinumab, and rilonacept, are used for the treatment of gout. Although the data are limited, these agents may provide a promising treatment option for acute gout in patients with contraindications to, or who are refractory to, traditional therapies like NSAIDs or colchicine. A recent study suggests that canakinumab, a fully human anti-IL-1β monoclonal antibody, can provide rapid and sustained pain relief at a dose of 150 mg subcutaneously. These medications are also being evaluated as therapies for prevention of gout flares while initiating urate-lowering therapy.

PREPARATIONS AVAILABLE



REFERENCES General Hellman DB, Imboden JB Jr: Arthritis and musculoskeletal disorders. In: McPhee ST , Papadakis MA (editors). Current Medical Diagnosis & Treatment, 2011. McGraw-Hill, 2011.

NSAIDs Chan FK et al: Celecoxib versus diclofenac and omeprazole in reducing the risk of recurrent ulcer bleeding in patients with arthritis. N Engl J Med 2002;347:2104. Clevers H: Colon cancer—Understanding how NSAIDs work. N Engl J Med 2006;354:761. Deeks JJ, Smith LA, Bradley MD: Efficacy, tolerability, and upper gastrointestinal safety of celecoxib for treatment of osteoarthritis and rheumatoid arthritis: Systematic review of randomised controlled trials. BMJ 2002;325:619. Furst DE et al: Dose response and safety study of meloxicam up to 22.5 mg daily in rheumatoid arthritis: A 12 week multi-center, double blind, dose response study versus placebo and diclofenac. J Rheumatol 2002;29:436. Knijff-Dutmer EA et al: Platelet function is inhibited by non-selective non-steroidal anti-inflammatory drugs but not by cyclooxygenase-2-selective inhibitors in patients with rheumatoid arthritis. Rheumatology (Oxford) 2002;41:458. Lago P et al: Safety and efficacy of ibuprofen versus indomethacin in preterm infants treated for patent ductus arteriosus: A randomized controlled trial. Eur J Pediatr 2002;161:202. Laine L et al: Serious lower gastrointestinal clinical events with non-selective NSAID or coxib use. Gastroenterology 2003;124:288. Moran EM: Epidemiological and clinical aspects of nonsteroidal anti-inflammatory drugs and cancer risks. J Environ Pathol T oxicol Oncol 2002;21:193. Niccoli L, Bellino S, Cantini F: Renal tolerability of three commonly employed non-steroidal anti-inflammatory drugs in elderly patients with osteoarthritis. Clin Exp Rheumatol 2002;20:201. Ray WA et al: COX-2 selective non-steroidal anti-inflammatory drugs and risk of serious coronary heart disease. Lancet 2002;360:1071. Rovensky J et al: T reatment of knee osteoarthritis with a topical nonsteroidal anti-inflammatory drug. Results of a randomized, double-blind, placebo-controlled study on the efficacy and safety of a 5% ibuprofen cream. Drugs Exp Clin Res 2001;27:209. Vane J, Botting R: Inflammation and the mechanism of action of anti-inflammatory drugs. FASEB J 1987;1:89. http://www.rheumatology.org/publications/hotline/0305NSAIDs.asp.

Disease-Modifying Antirheumatic Drugs & Glucocorticoids Atzeni F et al: Potential target of infliximab in autoimmune and inflammatory diseases. Autoimmun Rev 2007;6:8. Bannwarth B, Kostine M, Poursac N: A pharmacokinetic and clinical assessment of tofacitinib for the treatment of rheumatoid arthritis. Expert Opin Drug Metab T oxicol 2013;9:6. Besada E, Koldingsnes W, Nossent J: Characteristics of late onset neutropenia in rheumatologic patients treated with rituximab: A case review analysis from a single center. QJM 2012;105:6. Bongartz T et al: Anti-T NF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies. JAMA 2006;295:2275. Conklyn M et al: T he JAK3 inhibitor CP-690550 selectively reduces NK and CD8+ cell numbers in cynomolgus monkey blood following chronic oral dosing. J Leukoc Biol 2004;76:6. Cronstein B: How does methotrexate suppress inflammation? Clin Exp Rheumatol 2010:28(Suppl 61):S21. Dinarello CA, Simon A, van der Meer JW: T reating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012;11:8. Emery P et al: Golimumab, a human anti-tumor necrosis factor α monoclonal antibody, injected subcutaneously every four weeks in methotrexate-naïve patients with active rheumatoid arthritis. Arthritis Rheum 2009;60(8):2272. Emery P et al: IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomized placebo-controlled trial. Ann Rheum Dis 2008;67:1516. Feagan BG et al: T he effects of infliximab therapy on health-related quality of life in ulcerative colitis patients. Am J Gastroenterol 2007;102:4. Furst DE: Rational use of disease-modifying antirheumatic drugs. Drugs 1990;39:19. Furst DE et al: Updated consensus statement on biological agents for the treatment of rheumatic diseases, 2012. Ann Rheum Dis 2013;72. Gabay C et al: T ocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACT A): A randomised, double-blind, controlled phase 4 trial. Lancet 2013;4(9877):381. Genovese MC et al: Abatacept for rheumatoid arthritis refractory to tumor necrosis factor α inhibition. N Engl J Med 2005;353:1114. Genovese MC et al: Subcutaneous abatacept versus intravenous abatacept: A phase IIIb noninferiority study in patients with an inadequate response to methotrexate. Arthritis Rheum 2011;63:10. Keystone E et al: Improvement in patient-reported outcomes in a rituximab trial in patients with severe rheumatoid arthritis refractory to anti-tumor necrosis factor therapy. Arthritis Rheum 2008;59:785. Kobayashi K et al: Leukoencephalopathy with cognitive impairment following tocilizumab for the treatment of rheumatoid arthritis (RA). Intern Med 2009;48:15. Kremer J: T oward a better understanding of methotrexate. Arthritis Rheum 2004;50:1370. Landewé R et al: Efficacy of certolizumab pegol on signs and symptoms of axial spondyloarthritis including ankylosing spondylitis: 24-week results of a double-blind randomised placebo-controlled Phase 3 study. Ann Rheum Dis 2014;73:1. Maurizio Cutolo: T he kinase inhibitor tofacitinib in patients with rheumatoid arthritis: Latest findings and clinical potential. T her Adv Musculoskelet Dis 2013;5:1. Mease PJ et al: Effect of certolizumab pegol on signs and symptoms in patients with psoriatic arthritis: 24-week results of a Phase 3 double-blind randomised placebocontrolled study (RAPID-PsA). Ann Rheum Dis 2014;73:1. Nadashkevich O et al: A randomized unblinded trial of cyclophosphamide versus azathioprine in the treatment of systemic sclerosis. Clin Rheumatol 2006;25.


Ørum M et al: Beneficial effect of infliximab on refractory sarcoidosis. Dan Med J 2012;59:12. Papoutsaki M et al: Infliximab in psoriasis and psoriatic arthritis. BioDrugs 2013;27 (Suppl 1):13. Plosker G, Croom K: Sulfasalazine: A review of its use in the management of rheumatoid arthritis. Drugs 2006;65:1825. Riese RJ, Krishnaswami S, Kremer J: Inhibition of JAK kinases in patients with rheumatoid arthritis: Scientific rationale and clinical outcomes. Best Pract Res Clin Rheumatol 2010;24:4. Ruperto N et al: Abatacept in children with juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled withdrawal trial. Lancet 2008;2(9636):372. Scott DL, Kingsley GH: T umor necrosis factor inhibitors for rheumatoid arthritis. N Engl J Med 2006;355:704. Smolen J et al: Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomized controlled trial. Ann Rheum Dis 2009;68:797. Spies CM et al: Prednisone chronotherapy. Clin Exp Rheumatol 2011;29 (Suppl 68):5. Strober B et al: Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol 2013;169:5. T anaka T , Ogata A, Narazaki M: T ocilizumab for the treatment of rheumatoid arthritis. Expert Rev Clin Immunol 2010;6:6. T eng GG, T urkiewicz AM, Moreland LW: Abatacept: A costimulatory inhibitor for treatment of rheumatoid arthritis. Expert Opin Biol T her 2005;5:1245. T urner D: Severe acute ulcerative colitis: the pediatric perspective. Dig Dis 2009;27:3 van Gurp EA et al: T he effect of the JAK inhibitor CP-690,550 on peripheral immune parameters in stable kidney allograft patients. T ransplantation 2009;87:1. Weinblatt M et al: Adalimumab, a fully human anti-tumor necrosis factor α monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate. Arthritis Rheum 2003;48(1):35. Weinblatt ME et al: Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: Findings of a phase IIIb, multinational, prospective, randomized study. Arthritis Rheum 2013;65:1. Yokota S, Kishimoto T : T ocilizumab: Molecular intervention therapy in children with systemic juvenile idiopathic arthritis. Expert Rev Clin Immunol 2010;6:5. Zouali M, Uy EA: Belimumab therapy in systemic lupus erythematosus. BioDrugs 2013;27:3.

Other Analgesics Chandrasekharan NV et al: COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression. Proc Natl Acad Sci USA 2002;99:13926. Lee CR, McT avish D, Sorkin EM: T ramadol. A preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in acute and chronic pain states. Drugs 1993;46:2.

Drugs Used in Gout Becker MA et al: Febuxostat compared with allopurinol in patients with hyperuricemia and gout. N Engl J Med 2005;353:2450. Getting SJ et al: Activation of melanocortin type 3 receptor as a molecular mechanism for adrenocorticotropic hormone efficacy in gouty arthritis. Arthritis Rheum 2002;46:2765. Schumacher HR: Febuxostat: A non-purine, selective inhibitor of xanthine oxidase for the management of hyperuricaemia in patients with gout. Expert Opin Investig Drugs 2005;14:893. So A et al: A pilot study of IL-1 inhibition by anakinra in acute gout. Arthritis Res T her 2007;9:R28. Wallace SL, Singer JZ: Systemic toxicity associated with intravenous administration of colchicine—Guidelines for use. J Rheumatol 1988;15:495. http://www.fda.gov/cder/drugs/unapproved_drugs/colchicine_qa.htm (Restriction on drugs containing colchicine)

CASE STUDY ANSWER This patient had good control of his symptoms for 1 year but now has a prolonged flare, probably denoting worsening disease (not just a temporary flare). In addition to physical findings and measurement of acute-phase reactants such as sedimentation rate or Creactive protein, it would be wise to get hand and feet radiographs to document whether he has developed joint damage. Assuming such damage is found, the appropriate approach would be either a combination of nonbiologic DMARDs (eg, adding sulfasalazine and hydroxychloroquine) or adding a biologic medication, usually a TNF inhibitor. Follow-up should be every 1–3 months to gauge response and toxicity. Adverse events requiring caution are an increased risk of infection, possible appearance of lymphoma and rare liver function test or hematologic abnormalities. Importantly, close follow-up should ensue, including changing medications every 3–6 months until full disease control is achieved.


_______________ * Listed alphabetically.


SECTION VII ENDOCRINE DRUGS


CHAPTER

37 Hypothalamic & Pituitary Hormones Roger K. Long, MD, & Hakan Cakmak, MD

CASE STUDY A 3-year-old boy (height 85 cm, –3 standard deviations [SD]; weight 13 kg, approximately 10th percentile) presents with short stature. Review of the past history and growth chart demonstrates normal birth weight and birth length, but a progressive fall off in height velocity relative to age-matched normal ranges starting at 6 months of age. Physical examination demonstrates short stature and mild generalized obesity. Genital examination reveals descended but small testes and a phallic length of –2 SD. Laboratory evaluations demonstrate growth hormone (GH) deficiency and a delayed bone age of 18 months. The patient is started on replacement with recombinant human GH at a dose of 40 mcg/kg/d subcutaneously. After 1 year of treatment, his height velocity has increased from 5 cm/y to 11 cm/y. How does GH stimulate growth in children? What other hormone deficiencies are suggested by the patient’s physical examination? What other hormone supplementation is this patient likely to require?

The control of metabolism, growth, and reproduction is mediated by a combination of neural and endocrine systems located in the hypothalamus and pituitary gland. The pituitary weighs about 0.6 g and rests at the base of the brain in the bony sella turcica near the optic chiasm and the cavernous sinuses. The pituitary consists of an anterior lobe (adenohypophysis) and a posterior lobe (neurohypophysis) (Figure 37–1). It is connected to the overlying hypothalamus by a stalk of neurosecretory fibers and blood vessels, including a portal venous system that drains the hypothalamus and perfuses the anterior pituitary. The portal venous system carries small regulatory hormones (Figure 37–1, Table 37–1) from the hypothalamus to the anterior pituitary.


FIGURE 37–1 The hypothalamic-pituitary endocrine system. Hormones released from the anterior pituitary stimulate the production of hormones by a peripheral endocrine gland, the liver, or other tissues, or act directly on target tissues. Prolactin and the hormones released from the posterior pituitary (vasopressin and oxytocin) act directly on target tissues. Hypothalamic factors regulate the release of anterior pituitary hormones. ACTH, adrenocorticotropin; ADH, antidiuretic hormone [vasopressin]; CRH, corticotropin-releasing hormone; DA, dopamine; FSH, follicle-stimulating hormone; GH, growth hormone; GHRH, growth hormone-releasing hormone; GnRH, gonadotropinreleasing hormone; LH, luteinizing hormone; PRL, prolactin; SST, somatostatin; TRH, thyrotropin-releasing hormone; TSH, thyroid-


stimulating hormone. TABLE 37–1 Links between hypothalamic, anterior pituitary, and target organ hormone or mediator.1

The posterior lobe hormones are synthesized in the hypothalamus and transported via the neurosecretory fibers in the stalk of the pituitary to the posterior lobe; from there they are released into the circulation. Drugs that mimic or block the effects of hypothalamic and pituitary hormones have pharmacologic applications in three primary areas: (1) as replacement therapy for hormone deficiency states; (2) as antagonists for diseases caused by excess production of pituitary hormones; and (3) as diagnostic tools for identifying several endocrine abnormalities.

ANTERIOR PITUITARY HORMONES & THEIR HYPOTHALAMIC REGULATORS All the hormones produced by the anterior pituitary except prolactin are key participants in hormonal systems in which they regulate the production of hormones and autocrine-paracrine factors by endocrine glands and other peripheral tissues. In these systems, the secretion of the pituitary hormone is under the control of one or more hypothalamic hormones. Each hypothalamic-pituitary-endocrine gland system or axis provides multiple opportunities for complex neuroendocrine regulation of growth and development, metabolism, and reproductive function.

ANTERIOR PITUITARY & HYPOTHALAMIC HORMONE RECEPTORS The anterior pituitary hormones can be classified according to hormone structure and the types of receptors that they activate. Growth hormone (GH) and prolactin (PRL), single-chain protein hormones with significant homology, form one group. Both hormones activate receptors of the JAK/STAT superfamily (see Chapter 2). Three pituitary hormones—thyroid-stimulating hormone (TSH, thyrotropin), follicle-stimulating hormone (FSH), and luteinizing hormone (LH)—are dimeric proteins that activate G proteincoupled receptors (see Chapter 2). TSH, FSH, and LH share a common α subunit. Their β subunits, though somewhat similar to each other, differ enough to confer receptor specificity. Finally, adrenocorticotropic hormone (ACTH), a single peptide cleaved from a larger precursor, pro-opiomelanocortin (POMC), that can be cleaved into various other biologically active peptides like α-melanocyte-


stimulating hormone (MSH) and β-endorphin (see Chapter 31), represents a third category. Like TSH, LH, and FSH, ACTH acts through a G protein-coupled receptor. A unique feature of the ACTH receptor (also known as the melanocortin 2 receptor) is that a transmembrane protein, melanocortin 2 receptor accessory protein, is essential for normal ACTH receptor trafficking and signaling. ACRONYMS

TSH, FSH, LH, and ACTH share similarities in the regulation of their release from the pituitary. Each is under the control of a distinctive hypothalamic peptide that stimulates their production by acting on G protein-coupled receptors (Table 37–1). TSH release is regulated by thyrotropin-releasing hormone (TRH), whereas the release of LH and FSH (known collectively as gonadotropins) is stimulated by pulses of gonadotropin-releasing hormone (GnRH). ACTH release is stimulated by corticotropin-releasing hormone (CRH). An important regulatory feature shared by these four structurally related hormones is that they and their hypothalamic releasing factors are subject to feedback inhibitory regulation by the hormones whose production they control. TSH and TRH production are inhibited by the two key thyroid hormones, thyroxine and triiodothyronine (see Chapter 38). Gonadotropin and GnRH production is inhibited in women by estrogen and progesterone, and in men by testosterone and other androgens. ACTH and CRH production are inhibited by cortisol. Feedback regulation is critical to the physiologic control of thyroid, adrenal cortical, and gonadal function and is also important in pharmacologic treatments that affect these systems. The hypothalamic hormonal control of GH and prolactin differs from the regulatory systems for TSH, FSH, LH, and ACTH. The hypothalamus secretes two hormones that regulate GH; growth hormone-releasing hormone (GHRH) stimulates GH production,


whereas the peptide somatostatin (SST) inhibits GH production. GH and its primary peripheral mediator, insulin-like growth factor-I (IGF-I), also provide feedback to inhibit GH release. Prolactin production is inhibited by the catecholamine dopamine acting through the D2 subtype of dopamine receptors. The hypothalamus does not produce a hormone that specifically stimulates prolactin secretion, although TRH can stimulate prolactin release, particularly when TRH concentrations are high in the setting of primary hypothyroidism. Whereas all the pituitary and hypothalamic hormones described previously are available for use in humans, only a few are of major clinical importance. Because of the greater ease of administration of target endocrine gland hormones or their synthetic analogs, the related hypothalamic and pituitary hormones (TRH, TSH, CRH, ACTH, GHRH) are used infrequently as treatments. Some, such as ACTH, are used for specialized diagnostic testing. These agents are described in Tables 37–2 and 37–3 and are not discussed further in this chapter. In contrast, GH, SST, LH, FSH, GnRH, and dopamine or analogs of these hormones are commonly used and are described in the following text. TABLE 37–2 Clinical uses of hypothalamic hormones and their analogs.

TABLE 37–3 Diagnostic uses of thyroid-stimulating hormone and adrenocorticotropin.


GROWTH HORMONE (SOMATOTROPIN) Growth hormone, an anterior pituitary hormone, is required during childhood and adolescence for attainment of normal adult size and has important effects throughout postnatal life on lipid and carbohydrate metabolism, and on lean body mass and bone density. Its growthpromoting effects are primarily mediated via IGF-I (also known as somatomedin C). Individuals with congenital or acquired deficiency of GH during childhood or adolescence fail to reach their midparental target adult height and have disproportionately increased body fat and decreased muscle mass. Adults with GH deficiency also have disproportionately low lean body mass.

Chemistry & Pharmacokinetics A. Structure Growth hormone is a 191-amino-acid peptide with two sulfhydryl bridges. Its structure closely resembles that of prolactin. In the past, medicinal GH was isolated from the pituitaries of human cadavers. However, this form of GH was found to be contaminated with prions that could cause Creutzfeldt-Jakob disease. For this reason, it is no longer used. Somatropin, the recombinant form of GH, has a 191amino-acid sequence that is identical with the predominant native form of human GH. B. Absorption, Metabolism, and Excretion Circulating endogenous GH has a half-life of approximately 20 minutes and is predominantly cleared by the liver. Recombinant human GH (rhGH) is administered subcutaneously 6–7 times per week. Peak levels occur in 2–4 hours and active blood levels persist for approximately 36 hours.

Pharmacodynamics Growth hormone mediates its effects via cell surface receptors of the JAK/STAT cytokine receptor superfamily. The hormone has two distinct GH receptor binding sites. Dimerization of two GH receptors is stimulated by a single GH molecule and activates signaling cascades mediated by receptor-associated JAK tyrosine kinases and STATs (see Chapter 2). The hormone has complex effects on growth, body composition, and carbohydrate, protein, and lipid metabolism. The growth-promoting effects are mediated principally, but not solely, through an increase in the production of IGF-I. Much of the circulating IGF-I is produced in the liver. Growth hormone also stimulates production of IGF-I in bone, cartilage, muscle, kidney, and other tissues, where it has autocrine or paracrine roles. It stimulates longitudinal bone growth until the epiphyses close—near the end of puberty. In both children and adults, GH has anabolic effects in muscle and catabolic effects in adipose cells that shift the balance of body mass to an increase in muscle mass and a reduction in adiposity. The direct and indirect effects of GH on carbohydrate metabolism are mixed, in part because GH and IGF-I have opposite effects on insulin sensitivity. Growth hormone reduces insulin sensitivity, which results in mild hyperinsulinemia and increased blood


glucose levels, whereas IGF-I has insulin-like effects on glucose transport. In patients who are unable to respond to growth hormone because of severe resistance (caused by GH receptor mutations, post- receptor signaling mutations, or GH antibodies), the administration of recombinant human IGF-I may cause hypoglycemia because of its insulin-like effects.

Clinical Pharmacology A. Growth Hormone Deficiency Growth hormone deficiency can have a genetic basis, be associated with midline developmental defect syndromes (eg, septo-optic dysplasia), or be acquired as a result of damage to the pituitary or hypothalamus by a traumatic event (including breech or traumatic delivery), intracranial tumors, infection, infiltrative or hemorrhagic processes, or irradiation. Neonates with isolated GH deficiency are typically of normal size at birth because prenatal growth is not GH-dependent. In contrast, IGF-I is essential for normal prenatal and postnatal growth. Through poorly understood mechanisms, IGF-I expression and postnatal growth become GH-dependent during the first year of life. In childhood, GH deficiency typically presents as short stature, often with mild adiposity. Another early sign of GH deficiency is hypoglycemia due to the loss of a counter-regulatory hormonal response to hypoglycemia; young children are at risk for this condition due to high sensitivity to insulin. Criteria for diagnosis of GH deficiency usually include (1) a subnormal height velocity for age and (2) a subnormal serum GH response following provocative testing with at least two GH secretagogues. Clonidine (ι2 -adrenergic agonist), levodopa (dopaminergic agonist), and exercise are factors that increase GHRH levels. Arginine and insulin-induced hypoglycemia cause diminished SST, which increases GH release. The prevalence of GH deficiency is approximately 1:5000. If therapy with rhGH is initiated at an early age, many children with short stature due to GH deficiency will achieve an adult height within their midparental target height range. In the past, it was believed that adults with GH deficiency do not exhibit a significant syndrome. However, more detailed studies suggest that adults with GH deficiency often have generalized obesity, reduced muscle mass, asthenia, diminished bone mineral density, dyslipidemia, and reduced cardiac output. Growth hormone-deficient adults who have been treated with GH experience reversal of many of these manifestations. B. Growth Hormone Treatment of Pediatric Patients with Short Stature Although the greatest improvement in growth occurs in patients with GH deficiency, exogenous GH has some effect on height in children with short stature caused by conditions other than GH deficiency. Growth hormone has been approved for several conditions (Table 37– 4) and has been used experimentally or off-label in many others. Prader-Willi syndrome is an autosomal dominant genetic disease associated with growth failure, obesity, and carbohydrate intolerance. In children with Prader-Willi syndrome and growth failure, GH treatment decreases body fat and increases lean body mass, linear growth, and energy expenditure. TABLE 37–4 Clinical uses of recombinant human growth hormone.


Growth hormone treatment has also been shown to have a strong beneficial effect on final height of girls with Turner syndrome (45 X karyotype and variants). In clinical trials, GH treatment has been shown to increase final height in girls with Turner syndrome by 10– 15 cm (4–6 inches). Because girls with Turner syndrome also have either absent or rudimentary ovaries, GH must be judiciously combined with gonadal steroids to achieve maximal height. Other conditions of pediatric growth failure for which GH treatment is approved include chronic renal insufficiency pre-transplant and small-for-gestational-age at birth in which the child’s height remains more than 2 standard deviations below normal at 2 years of age. A controversial but approved use of GH is for children with idiopathic short stature (ISS). This is a heterogeneous population that


has in common no identifiable cause of the short stature. Some have arbitrarily defined ISS clinically as having a height at least 2.25 standard deviations below normal for children of the same age and a predicted adult height that is less than 2.25 standard deviations below normal. In this group of children, many years of GH therapy result in an average increase in adult height of 4–7 cm (1.57–2.76 inches) at a cost of $5000–$40,000 per year. The complex issues involved in the cost-risk-benefit relationship of this use of GH are important because an estimated 400,000 children in the United States fit the diagnostic criteria for ISS. Treatment of children with short stature should be carried out by specialists experienced in GH administration. Dose requirements vary with the condition being treated, with GH-deficient children typically being most responsive. Children must be observed closely for slowing of growth velocity, which could indicate a need to increase the dosage or the possibility of epiphyseal fusion or intercurrent problems such as hypothyroidism or malnutrition.

Other Uses of Growth Hormone Growth hormone affects many organ systems and also has a net anabolic effect. It has been tested in a number of conditions that are associated with a severe catabolic state and is approved for the treatment of wasting in patients with AIDS. In 2004, GH was approved for treatment of patients with short bowel syndrome who are dependent on total parenteral nutrition (TPN). After intestinal resection or bypass, the remaining functional intestine in many patients undergoes extensive adaptation that allows it to adequately absorb nutrients. However, other patients fail to adequately adapt and develop a malabsorption syndrome. Growth hormone has been shown to increase intestinal growth and improve its function in experimental animals. Benefits of GH treatment for patients with short bowel syndrome and dependence on TPN have mostly been short-lived in the clinical studies that have been published to date. Growth hormone is administered with glutamine, which also has trophic effects on the intestinal mucosa. Growth hormone is a popular component of “anti-aging” programs. Serum levels of GH normally decline with aging; anti-aging programs claim that injection of GH or administration of drugs purported to increase GH release are effective anti-aging remedies. These claims are largely unsubstantiated. In contrast, studies in mice and the nematode Caenorhabditis elegans have clearly demonstrated that analogs of human GH and IGF-I consistently shorten life span and that loss-of-function mutations in the signaling pathways for the GH and IGF-I analogs lengthen life span. Another use of GH is by athletes for a purported increase in muscle mass and athletic performance. Growth hormone is one of the drugs banned by the International Olympic Committee. In 1993, the FDA approved the use of recombinant bovine growth hormone (rbGH) in dairy cattle to increase milk production. Although milk and meat from rbGH-treated cows appear to be safe, these cows have a higher incidence of mastitis, which could increase antibiotic use and result in greater antibiotic residues in milk and meat.

Toxicity & Contraindications Children generally tolerate growth hormone treatment well. Adverse events are relatively rare and include pseudotumor cerebri, slipped capital femoral epiphysis, progression of scoliosis, edema, hyperglycemia, and increased risk of asphyxiation in severely obese patients with Prader-Willi syndrome and upper airway obstruction or sleep apnea. Patients with Turner syndrome have an increased risk of otitis media while taking GH. In children with GH deficiency, periodic evaluation of the other anterior pituitary hormones may reveal concurrent deficiencies, which also require treatment (ie, with hydrocortisone, levothyroxine, or gonadal hormones). Pancreatitis, gynecomastia, and nevus growth have occurred in patients receiving GH. Adults tend to have more adverse effects from GH therapy. Peripheral edema, myalgias, and arthralgias (especially in the hands and wrists) occur commonly but remit with dosage reduction. Carpal tunnel syndrome can occur. Growth hormone treatment increases the activity of cytochrome P450 isoforms, which may reduce the serum levels of drugs metabolized by that enzyme system (see Chapter 4). There has been no increased incidence of malignancy among patients receiving GH therapy, but such treatment is contraindicated in a patient with a known active malignancy. Proliferative retinopathy may rarely occur. Growth hormone treatment of critically ill patients appears to increase mortality. The long-term health effects of GH treatment in childhood are unknown. The preliminary results from the Safety and Appropriateness of GH in Europe (SAGHE) study are variable. A higher all-cause mortality (mostly due to cardiovascular disease) was found in the GH treatment group in the French arm of the study, but no long-term risks of GH treatment were observed in the study arm from another region of Europe.

MECASERMIN A small number of children with growth failure have severe IGF-I deficiency that is not responsive to exogenous GH. Causes include mutations in the GH receptor and in the GH receptor signaling pathway, neutralizing antibodies to GH, and IGF-I gene defects. In 2005, the FDA approved two forms of recombinant human IGF-I (rhIGF-I) for treatment of severe IGF-I deficiency that is not responsive to GH: mecasermin and mecasermin rinfabate. Mecasermin is rhIGF-I alone, while mecasermin rinfabate is a complex of rhIGF-I and recombinant human insulin-like growth factor-binding protein-3 (rhIGFBP-3). This binding protein significantly increases the circulating half-life of rhIGF-I. Normally, the great majority of the circulating IGF-I is bound to IGFBP-3, which is produced principally by the liver under the control of GH. Mecasermin rinfabate is not currently available in the United States. Mecasermin is administered subcutaneously twice daily at a recommended starting dosage of 0.04–0.08 mg/kg and increased weekly up to a maximum twice-daily


dosage of 0.12 mg/kg. The most important adverse effect observed with mecasermin is hypoglycemia. To avoid hypoglycemia, the prescribing instructions require consumption of a carbohydrate-containing meal or snack 20 minutes before or after mecasermin administration. Several patients have experienced intracranial hypertension, adenotonsillar hypertrophy, and asymptomatic elevation of liver enzymes.

GROWTH HORMONE ANTAGONISTS Antagonists of GH are used to reverse the effects of GH-producing cells (somatotrophs) in the anterior pituitary that tend to form GHsecreting tumors. Hormone-secreting pituitary adenomas occur most commonly in adults. In adults, GH-secreting adenomas cause acromegaly, which is characterized by abnormal growth of cartilage and bone tissue, and many organs including skin, muscle, heart, liver, and the gastrointestinal tract. Acromegaly adversely affects the skeletal, muscular, cardiovascular, respiratory, and metabolic systems. When a GH-secreting adenoma occurs before the long bone epiphyses close, it leads to the rare condition, gigantism. Larger pituitary adenomas produce greater amounts of GH and also can impair visual and central nervous system function by encroaching on nearby brain structures. The initial therapy of choice for GH-secreting adenomas is transsphenoidal surgery. Medical therapy with GH antagonists is introduced if GH hypersecretion persists after surgery. These agents include somatostatin analogs and dopamine receptor agonists, which reduce the production of GH, and the novel GH receptor antagonist pegvisomant, which prevents GH from activating GH signaling pathways. Radiation therapy is reserved for patients with inadequate response to surgical and medical therapies.

Somatostatin Analogs Somatostatin, a 14-amino-acid peptide (Figure 37–2), is found in the hypothalamus, other parts of the central nervous system, the pancreas, and other sites in the gastrointestinal tract. It functions primarily as an inhibitory paracrine factor and inhibits the release of GH, TSH, glucagon, insulin, and gastrin. Somatostatin is rapidly cleared from the circulation, with a half-life of 1–3 minutes. The kidney appears to play an important role in its metabolism and excretion.

FIGURE 37–2 Above: Amino acid sequence of somatostatin. Below: Sequence of the synthetic analog, octreotide. Somatostatin has limited therapeutic usefulness because of its short duration of action and multiple effects in many secretory systems. A series of longer-acting somatostatin analogs that retain biologic activity have been developed. Octreotide, the most widely used somatostatin analog (Figure 37–2), is 45 times more potent than somatostatin in inhibiting GH release but only twice as potent in reducing insulin secretion. Because of this relatively reduced effect on pancreatic beta cells, hyperglycemia rarely occurs during treatment. The plasma elimination half-life of octreotide is about 80 minutes, 30 times longer than that of somatostatin. Octreotide, 50–200 mcg given subcutaneously every 8 hours, reduces symptoms caused by a variety of hormone-secreting tumors: acromegaly, carcinoid syndrome, gastrinoma, glucagonoma, insulinoma, VIPoma, and ACTH-secreting tumor. Other therapeutic use indications include diarrhea—secretory, HIV associated, diabetic, chemotherapy, or radiation induced—and portal hypertension. Somatostatin receptor scintigraphy, using radiolabeled octreotide, is useful in localizing neuroendocrine tumors having somatostatin receptors and helps predict the response to octreotide therapy. Octreotide is also useful for the acute control of bleeding from esophageal varices.


Octreotide acetate injectable long-acting suspension is a slow-release microsphere formulation. It is instituted only after a brief course of shorter-acting octreotide has been demonstrated to be effective and tolerated. Injections into alternate gluteal muscles are repeated at 4-week intervals in doses of 10–40 mg. Adverse effects of octreotide therapy include nausea, vomiting, abdominal cramps, flatulence, and steatorrhea with bulky bowel movements. Biliary sludge and gallstones may occur after 6 months of use in 20–30% of patients. However, the yearly incidence of symptomatic gallstones is about 1%. Cardiac effects include sinus bradycardia (25%) and conduction disturbances (10%). Pain at the site of injection is common, especially with the long-acting octreotide suspension. Vitamin B 12 deficiency may occur with long-term use of octreotide. A long-acting formulation of lanreotide, another octapeptide somatostatin analog, is approved for treatment of acromegaly. Lanreotide appears to have effects comparable to those of octreotide in reducing GH levels and normalizing IGF-I concentrations.

Pegvisomant Pegvisomant is a GH receptor antagonist used to treat acromegaly. It is the polyethylene glycol (PEG) derivative of a mutant GH, B2036. Pegylation reduces its clearance and improves its overall clinical effectiveness. Like native GH, pegvisomant has two GH receptor binding sites. However, one of its GH receptor binding sites has increased affinity for the GH receptor, whereas its second GH receptor binding site has reduced affinity. This differential receptor affinity allows the initial step (GH receptor dimerization) but blocks the conformational changes required for signal transduction. In clinical trials, pegvisomant was administered subcutaneously to patients with acromegaly; daily treatment for 12 months or more reduced serum levels of IGF-I into the normal range in 97%. Pegvisomant does not inhibit GH secretion and may lead to increased GH levels and possible adenoma growth. No serious problems have been observed; however, increases in liver enzymes without liver failure have been reported.

THE GONADOTROPINS (FOLLICLE-STIMULATING HORMONE & LUTEINIZING HORMONE) & HUMAN CHORIONIC GONADOTROPIN The gonadotropins are produced by gonadotroph cells, which comprise 7–15% of the cells in the pituitary. These hormones serve complementary functions in the reproductive process. In women, the principal function of FSH is to stimulate ovarian follicle development. Both FSH and LH are needed for ovarian steroidogenesis. In the ovary, LH stimulates androgen production by theca cells in the follicular stage of the menstrual cycle, whereas FSH stimulates the conversion of androgens to estrogens by granulosa cells. In the luteal phase of the menstrual cycle, estrogen and progesterone production is primarily under the control first of LH and then, if pregnancy occurs, under the control of human chorionic gonadotropin (hCG). Human chorionic gonadotropin is a placental glycoprotein nearly identical with LH; its actions are mediated through LH receptors. In men, FSH is the primary regulator of spermatogenesis, whereas LH is the main stimulus for testosterone synthesis in Leydig cells. FSH helps maintain high local androgen concentrations in the vicinity of developing sperm by stimulating the production of androgenbinding protein in Sertoli cells. FSH also stimulates the conversion by Sertoli cells of testosterone to estrogen that is also required for spermatogenesis. FSH, LH, and hCG are available in several pharmaceutical forms. They are used in states of infertility to stimulate spermatogenesis in men and to induce follicle development and ovulation in women. Their most common clinical use is for the controlled ovarian stimulation that is the cornerstone of assisted reproductive technologies such as in vitro fertilization (IVF, see below).

Chemistry & Pharmacokinetics All three hormones—FSH, LH, and hCG—are heterodimers that share an identical α subunit in addition to a distinct β subunit that confers receptor specificity. The β subunits of hCG and LH are nearly identical, and these two hormones are used interchangeably. All the gonadotropin preparations are administered by subcutaneous or intramuscular injection, usually on a daily basis. Half-lives vary by preparation and route of injection from 10 to 40 hours. A. Menotropins The first commercial gonadotropin product containing both FSH and LH was extracted from the urine of postmenopausal women. This purified extract of FSH and LH is known as menotropins, or human menopausal gonadotropins (hMG). From the early 1960s, these preparations were used for the stimulation of follicle development in women. The early extraction techniques were very crude, requiring around 30 L of urine to manufacture enough hMG needed for a single treatment cycle. These initial preparations were also contaminated with other proteins; less than 5% of the proteins present were bioactive. The FSH-to-LH bioactivity ratio of these early preparations was 1:1. As purity improved, it was necessary to add hCG in order to maintain this ratio of bioactivity. B. Follicle-Stimulating Hormone


Three forms of purified FSH are available. Urofollitropin, also known as uFSH, is a purified preparation of human FSH extracted from the urine of postmenopausal women. Virtually all the LH activity has been removed through a form of immuno-affinity chromatography that uses anti-hCG antibodies. Two recombinant forms of FSH (rFSH) are also available: follitropin alfa and follitropin beta. The amino acid sequences of these two products are identical to that of human FSH. They differ from each other and urofollitropin in the composition of carbohydrate side chains. The rFSH preparations have a shorter half-life than preparations derived from human urine but stimulate estrogen secretion at least as efficiently and, in some studies, more efficiently. Compared with urine derived gonadotropins, rFSH preparations have little protein contamination, much less batch-to-batch variability, and may cause less local tissue reaction. The rFSH preparations are considerably more expensive. C. Luteinizing Hormone Lutropin alfa, the first and only recombinant form of human LH, was introduced in the United States in 2004. When given by subcutaneous injection, it has a half-life of about 10 hours. Lutropin has only been approved for use in combination with follitropin alfa for stimulation of follicular development in infertile hypogonadotropic hypogonadal women with profound LH deficiency (< 1.2 IU/L). Lutropin alfa with follitropin alfa may also be of benefit in certain subgroups of normogonadotropic women (eg, those with an inadequate response to prior follitropin alfa monotherapy). It has not been approved for use with the other preparations of FSH or for induction of ovulation. Lutropin alfa was withdrawn from the U.S. market in 2012. D. Human Chorionic Gonadotropin Human chorionic gonadotropin is produced by the human placenta and excreted into the urine, whence it can be extracted and purified. It is a glycoprotein consisting of a 92-amino-acid α subunit virtually identical to that of FSH, LH, and TSH, and a β subunit of 145 amino acids that resembles that of LH except for the presence of a carboxyl terminal sequence of 30 amino acids not present in LH. Choriogonadotropin alfa (rhCG) is a recombinant form of hCG. Because of its greater consistency in biologic activity, rhCG is packaged and dosed on the basis of weight rather than units of activity. All of the other gonadotropins, including rFSH, are packaged and dosed on the basis of units of activity. Both the hCG preparation that is purified from human urine and rhCG can be administered by subcutaneous or intramuscular injection.

Pharmacodynamics The gonadotropins and hCG exert their effects through G protein-coupled receptors. LH and FSH have complex effects on reproductive tissues in both sexes. In women, these effects change over the time course of a menstrual cycle as a result of a complex interplay among concentration-dependent effects of the gonadotropins, cross-talk of LH, FSH, and gonadal steroids, and the influence of other ovarian hormones. A coordinated pattern of FSH and LH secretion during the menstrual cycle (see Figure 40–1) is required for normal follicle development, ovulation, and pregnancy. During the first 8 weeks of pregnancy, the progesterone and estrogen required to maintain pregnancy are produced by the ovarian corpus luteum. For the first few days after ovulation, the corpus luteum is maintained by maternal LH. However, as maternal LH concentrations fall owing to increasing concentrations of progesterone and estrogen, the corpus luteum will continue to function only if the role of maternal LH is taken over by hCG produced by syncytiotrophoblast cells in the placenta.

Clinical Pharmacology A. Ovulation Induction The gonadotropins are used to induce follicle development and ovulation in women with anovulation that is secondary to hypogonadotropic hypogonadism, polycystic ovary syndrome, obesity, and other causes. Because of the high cost of gonadotropins and the need for close monitoring during their administration, they are generally reserved for anovulatory women who fail to respond to other less complicated forms of treatment (eg, clomiphene; see Chapter 40). Gonadotropins are also used for controlled ovarian stimulation in assisted reproductive technology procedures. Currently, a number of different protocols use gonadotropins in ovulation induction and controlled ovulation stimulation, and new protocols are continually being developed to improve the rates of success and to decrease the two primary risks of ovulation induction: multiple pregnancies and the ovarian hyperstimulation syndrome (OHSS; see below). Although the details differ, all of these protocols are based on the complex physiology that underlies a normal menstrual cycle. Like a menstrual cycle, ovulation induction is discussed in relation to a cycle that begins on the first day of a menstrual bleed (Figure 37–3). Shortly after the first day (usually on day 2), daily injections with one of the FSH preparations (hMG, urofollitropin, or rFSH) are begun and continued for approximately 7–12 days. In women with hypogonadotropic hypogonadism, follicle development requires treatment with a combination of FSH and LH because these women do not produce the basal level of LH that is required for normal follicle development. The dose and duration of gonadotropin treatment are based on the response as measured by the serum estradiol concentration and by ultrasound evaluation of ovarian follicle development. When exogenous gonadotropins are used to stimulate follicle development, there is risk of a premature endogenous surge in LH owing to the rapidly increasing serum estradiol levels. To prevent this,


gonadotropins are almost always administered in conjunction with a drug that blocks the effects of endogenous GnRH—either continuous administration of a GnRH agonist, which down-regulates GnRH receptors or a GnRH receptor antagonist (see below and Figure 37–3).

FIGURE 37–3 Controlled ovarian stimulation in preparation for an assisted reproductive technology such as in vitro fertilization. Follicular phase: Follicle development is stimulated with gonadotropin injections that begin about 2 days after menses begin. When the follicles are ready, as assessed by ultrasound measurement of follicle size, final oocyte maturation is induced by an injection of hCG. Luteal phase: Shortly thereafter oocytes are retrieved and fertilized in vitro. The recipient’s luteal phase is supported with injections of progesterone. To prevent a premature luteinizing-hormone surge, endogenous LH secretion is inhibited with either a GnRH agonist or a GnRH antagonist. In most protocols, the GnRH agonist is started midway through the preceding luteal cycle. When appropriate follicular maturation has occurred, the gonadotropin and the GnRH agonist or GnRH antagonist injections are discontinued and hCG (3300–10,000 IU) is administered subcutaneously to induce final follicular maturation and, in ovulation induction protocols, ovulation. The hCG administration is followed by insemination in ovulation induction and by oocyte retrieval in assisted reproductive technology procedures. Because use of GnRH agonists or antagonists during the follicular phase of ovulation induction suppresses endogenous LH production, it is important to provide exogenous hormonal support of the luteal phase. In clinical trials, exogenous progesterone, hCG, or a combination of the two have been effective at providing adequate luteal support. However, progesterone is preferred for luteal support because hCG carries a higher risk of OHSS in patients with high follicular response to gonadotropins. B. Male Infertility Most of the signs and symptoms of hypogonadism in males (eg, delayed puberty, retention of prepubertal secondary sex characteristics after puberty) can be adequately treated with exogenous androgen; however, treatment of infertility in hypogonadal men requires the activity of both LH and FSH. For many years, conventional therapy has consisted of initial treatment for 8–12 weeks with injections of 1000–2500 IU hCG several times per week. After the initial phase, hMG is injected at a dose of 75–150 units three times per week. In men with hypogonadal hypogonadism, it takes an average of 4–6 months of such treatment for sperm to appear in the ejaculate in up to 90% of patients, but often not at normal levels. Even if pregnancy does not occur spontaneously, the number of sperm is often sufficient that pregnancy can be achieved by insemination with the patient’s semen (intrauterine insemination) or with the help of an assisted reproductive technique such as in vitro fertilization with or without intracytoplasmic sperm injection (ICSI), in which a single sperm is injected directly into a mature oocyte that has been retrieved after controlled ovarian stimulation of a female partner. With the advent of ICSI, the minimum threshold of spermatogenesis required for pregnancy is greatly lowered. C. Outdated Uses


Chorionic gonadotropin is approved for the treatment of prepubertal cryptorchidism. Prepubertal boys were treated with intramuscular injections of hCG for 2–6 weeks. However, this clinical use is no longer supported because the long-term efficacy of hormonal treatment of cryptorchidism (~ 20%) is much lower than the long-term efficacy of surgical treatment (> 95%), and because of concerns that early childhood treatment with hCG treatment has a negative impact on germ cells in addition to increasing the risk of precocious puberty. In the United States, chorionic gonadotropin has a black-box warning against its use for weight loss. The use of hCG plus severe calorie restriction for weight loss was popularized by a publication in the 1950s claiming that the hCG selectively mobilizes body fat stores. This practice continues today, despite a preponderance of subsequent scientific evidence from placebo-controlled trials that hCG does not provide any weight loss benefit beyond the weight loss associated with severe calorie restriction alone.

Toxicity & Contraindications In women treated with gonadotropins and hCG, the two most serious complications are OHSS and multiple pregnancies. Stimulation of the ovary during ovulation induction often leads to uncomplicated ovarian enlargement that usually resolves spontaneously. However, OHSS may occur and can be associated with ovarian enlargement, intravascular depletion, ascites, liver dysfunction, pulmonary edema, electrolyte imbalance, and thromboembolic events. Although OHSS is often self-limited, with spontaneous resolution within a few days, severe disease may require hospitalization and intensive care. Triggering the final oocyte maturation with hCG carries the risk of inducing OHSS. GnRH agonists also induce this final oocyte maturation by promoting the release of endogenous gonadotropin stores from the hypophysis and can be used as an alternative to hCG. Use of the GnRH agonist trigger dramatically reduces the risk of OHSS, owing to the short half-life of the GnRH agonist-induced endogenous LH surge. The probability of multiple pregnancies is greatly increased when ovulation induction and assisted reproductive technologies are used. In ovulation induction, the risk of a multiple pregnancy is estimated to be 5–10%, whereas the percentage of multiple pregnancies in the general population is closer to 1%. Multiple pregnancies carry an increased risk of complications, such as gestational diabetes, preeclampsia, and preterm labor. For in vitro fertilization procedures, the risk of a multiple pregnancy is primarily determined by the number of embryos transferred to the recipient. A strong trend in recent years has been to transfer single embryos. Other reported adverse effects of gonadotropin treatment are headache, depression, edema, precocious puberty, and (rarely) production of antibodies to hCG. In men treated with gonadotropins, the risk of gynecomastia is directly correlated with the level of testosterone produced in response to treatment. An association between ovarian cancer, infertility, and fertility drugs has been reported. However, it is not known which, if any, fertility drugs are causally related to cancer.

GONADOTROPIN-RELEASING HORMONE & ITS ANALOGS Gonadotropin-releasing hormone is secreted by neurons in the hypothalamus. It travels through the hypothalamic-pituitary venous portal plexus to the anterior pituitary, where it binds to G protein-coupled receptors on the plasma membranes of gonadotrophs. Pulsatile GnRH secretion is required to stimulate the gonadotrophs to produce and release LH and FSH. Sustained nonpulsatile administration of GnRH or GnRH analogs inhibits the release of FSH and LH by the pituitary in both women and men, resulting in hypogonadotropic hypogonadism. GnRH agonists are used to induce gonadal suppression in men with prostate cancer or children with central precocious puberty. They are also used in women who are undergoing assisted reproductive technology procedures or who have a gynecologic problem that is benefited by ovarian suppression.

Chemistry & Pharmacokinetics A. Structure GnRH is a decapeptide found in all mammals. Gonadorelin is an acetate salt of synthetic human GnRH. Substitution of amino acids at the 6 position or replacement of the C-terminal glycine-amide produces synthetic agonists. Both modifications make them more potent and longer-lasting than native GnRH and gonadorelin. Such analogs of GnRH include goserelin, buserelin, histrelin, leuprolide, nafarelin, and triptorelin. B. Pharmacokinetics Gonadorelin can be administered intravenously or subcutaneously. GnRH agonists can be administered subcutaneously, intramuscularly, via nasal spray (nafarelin), or as a subcutaneous implant. The half-life of intravenous gonadorelin is 4 minutes, and the half-lives of subcutaneous and intranasal GnRH analogs are approximately 3 hours. The duration of clinical uses of GnRH agonists varies from a few days for controlled ovarian stimulation to a number of years for treatment of metastatic prostate cancer. Therefore, preparations have been developed with a range of durations of action from several hours (for daily administration) to 1, 4, 6, or 12 months (depot forms).

Pharmacodynamics


The physiologic actions of GnRH exhibit complex dose-response relationships that change dramatically from the fetal period through the end of puberty. This is not surprising in view of the complex role that GnRH plays in normal reproduction, particularly in female reproduction. Pulsatile GnRH release occurs and is responsible for stimulating LH and FSH production during the fetal and neonatal period. Subsequently, from the age of 2 years until the onset of puberty, GnRH secretion falls off and the pituitary simultaneously exhibits very low sensitivity to GnRH. Just before puberty, an increase in the frequency and amplitude of GnRH release occurs and then, in early puberty, pituitary sensitivity to GnRH increases, which is due in part to the effect of increasing concentrations of gonadal steroids. In females, it usually takes several months to a year after the onset of puberty for the hypothalamic-pituitary system to produce an LH surge and ovulation. By the end of puberty, the system is well established so that menstrual cycles proceed at relatively constant intervals. The amplitude and frequency of GnRH pulses vary in a regular pattern through the menstrual cycle with the highest amplitudes occurring during the luteal phase and the highest frequency occurring late in the follicular phase. Lower pulse frequencies favor FSH secretion, whereas higher pulse frequencies favor LH secretion. Gonadal steroids as well as the peptide hormones activin, inhibin, and follistatin have complex modulatory effects on the gonadotropin response to GnRH. In the pharmacologic use of GnRH and its analogs, pulsatile intravenous administration of gonadorelin every 1–4 hours stimulates FSH and LH secretion. Continuous administration of gonadorelin or its longer-acting analogs produces a biphasic response. During the first 7–10 days, an agonist effect results in increased concentrations of gonadal hormones in males and females; this initial phase is referred to as a flare. After this period, the continued presence of GnRH results in an inhibitory action that manifests as a drop in the concentration of gonadotropins and gonadal steroids (ie, hypogonadotropic hypogonadal state). The inhibitory action is due to a combination of receptor down-regulation and changes in the signaling pathways activated by GnRH.

Clinical Pharmacology The GnRH agonists are occasionally used for stimulation of gonadotropin production. They are used far more commonly for suppression of gonadotropin release. A. Stimulation 1. Female infertility—In the current era of widespread availability of gonadotropins and assisted reproductive technology, the use of pulsatile GnRH administration to treat infertility is uncommon. Although pulsatile GnRH is less likely than gonadotropins to cause multiple pregnancies and OHSS, the inconvenience and cost associated with continuous use of an intravenous pump and difficulties obtaining native GnRH (gonadorelin) are barriers to pulsatile GnRH. When this approach is used, a portable battery-powered programmable pump and intravenous tubing deliver pulses of gonadorelin every 90 minutes. Gonadorelin or a GnRH agonist analog can be used to initiate an LH surge and ovulation in women with infertility who are undergoing ovulation induction with gonadotropins. Traditionally, hCG has been used to initiate ovulation in this situation. However, there is some evidence that gonadorelin or a GnRH agonist is less likely than hCG to cause OHSS. 2. Male infertility—It is possible to use pulsatile gonadorelin for infertility in men with hypothalamic hypogonadotropic hypogonadism. A portable pump infuses gonadorelin intravenously every 90 minutes. Serum testosterone levels and semen analyses must be done regularly. At least 3–6 months of pulsatile infusions are required before significant numbers of sperm are seen. As described above, treatment of hypogonadotropic hypogonadism is more commonly done with hCG and hMG or their recombinant equivalents. 3. Diagnosis of LH responsiveness—GnRH may be useful in determining whether delayed puberty in a hypogonadotropic adolescent is due to constitutional delay or to hypogonadotropic hypogonadism. The LH response (but not the FSH response) to a single dose of GnRH may distinguish between these two conditions; however, there can be significant individual overlap in the LH response between the two groups. Serum LH levels are measured before and at various times after an intravenous or subcutaneous bolus of GnRH. An increase in serum LH with a peak that is greater than 5–8 mIU/mL suggests early pubertal status. An impaired LH response suggests hypogonadotropic hypogonadism due to either pituitary or hypothalamic disease, but does not rule out constitutional delay of puberty. B. Suppression of Gonadotropin Production 1. Controlled ovarian stimulation—In the controlled ovarian stimulation that provides multiple mature oocytes for assisted reproductive technologies such as in vitro fertilization, it is critical to suppress an endogenous LH surge that could prematurely trigger ovulation. This suppression is most commonly achieved by daily subcutaneous injections of leuprolide or daily nasal applications of nafarelin. For leuprolide, treatment is commonly initiated with 1 mg daily for about 10 days until menstrual bleeding occurs. At that point, the dose is reduced to 0.5 mg daily until hCG is administered (Figure 37–3). For nafarelin, the beginning dosage is generally 400 mcg twice a day, which is decreased to 200 mcg when menstrual bleeding occurs. In women who respond poorly to the standard protocol, alternative protocols that use shorter courses may improve the follicular response to gonadotropins. 2. Endometriosis—Endometriosis is defined as the presence of estrogen-sensitive endometrium outside the uterus that results in cyclical abdominal pain in premenopausal women. The pain of endometriosis is often reduced by abolishing exposure to the cyclical changes in


the concentrations of estrogen and progesterone that are a normal part of the menstrual cycle. The ovarian suppression induced by continuous treatment with a GnRH agonist greatly reduces estrogen and progesterone concentrations and prevents cyclical changes. The preferred duration of treatment with a GnRH agonist is limited to 6 months because ovarian suppression beyond this period can result in decreased bone mineral density. When relief of pain from treatment with a GnRH agonist supports continued therapy for more than 6 months, the addition of add-back therapy (estrogen or progestins) reduces or eliminates GnRH agonist-induced bone mineral loss and provides symptomatic relief without reducing the efficacy of pain relief. Leuprolide and goserelin are administered as depot preparations that provide 1 or 3 months of continuous GnRH agonist activity. Nafarelin is administered twice daily as a nasal spray at a dose of 0.2 mg per spray. 3. Uterine leiomyomata (uterine fibroids)—Uterine leiomyomata are benign, estrogen-sensitive, smooth muscle tumors in the uterus that can cause menorrhagia, with associated anemia and pelvic pain. Treatment for 3–6 months with a GnRH agonist reduces fibroid size and, when combined with supplemental iron, improves anemia. The effects of GnRH agonists are temporary, with gradual recurrent growth of leiomyomas to previous size within several months after cessation of treatment. GnRH agonists have been used widely for preoperative treatment of uterine leiomyomas, both for myomectomy and hysterectomy. GnRH agonists have been shown to improve hematologic parameters, shorten hospital stay, and decrease blood loss, operating time, and postoperative pain when given for 3 months preoperatively. 4. Prostate cancer—Androgen deprivation therapy is the primary medical therapy for prostate cancer. Combined antiandrogen therapy with continuous GnRH agonist and an androgen receptor antagonist is as effective as surgical castration in reducing serum testosterone concentrations and effects. Leuprolide, goserelin, histrelin, buserelin, and triptorelin are approved for this indication. The preferred formulation is one of the long-acting depot forms that provide 1, 3, 4, 6, or 12 months of active drug therapy. During the first 7–10 days of GnRH analog therapy, serum testosterone levels increase because of the agonist action of the drug; this can precipitate pain in patients with bone metastases, and tumor growth and neurologic symptoms in patients with vertebral metastases. It can also temporarily worsen symptoms of urinary obstruction. Such tumor flares can usually be avoided with the concomitant administration of an androgen receptor antagonist (flutamide, bicalutamide, or nilutamide) (see Chapter 40). Within about 2 weeks, serum testosterone levels fall to the hypogonadal range. 5. Central precocious puberty—Continuous administration of a GnRH agonist is indicated for treatment of central precocious puberty (onset of secondary sex characteristics before 7–8 years in girls or 9 years in boys). Before embarking on treatment with a GnRH agonist, one must confirm central precocious puberty by demonstrating a pubertal gonadotropin response to GnRH or a “test dose” of a GnRH analog. Treatment is typically indicated in a child whose final height would be otherwise significantly compromised (as evidenced by a significantly advanced bone age) or in whom the early development of pubertal secondary sexual characteristics or menses causes significant emotional distress. While central precocious puberty is most often idiopathic, it is important to rule out central nervous system pathology with MRI imaging of the hypothalamic-pituitary area. Treatment is most commonly carried out with either a monthly or three-monthly intramuscular depot injection of leuprolide acetate or with a once-yearly implant of histrelin acetate. Daily subcutaneous regimens and multiple daily nasal spray regimens of GnRH agonists are also available. Treatment with a GnRH agonist is generally continued to age 11 in females and age 12 in males. 6. Other—The gonadal suppression provided by continuous GnRH agonist treatment is used in the management of advanced breast and ovarian cancer. In addition, recently published clinical practice guidelines recommend the use of continuous GnRH agonist administration in early pubertal transgender adolescents to block endogenous puberty prior to subsequent treatment with cross-gender gonadal hormones.

Toxicity Gonadorelin can cause headache, light-headedness, nausea, and flushing. Local swelling often occurs at subcutaneous injection sites. Generalized hypersensitivity dermatitis has occurred after long-term subcutaneous administration. Rare acute hypersensitivity reactions include bronchospasm and anaphylaxis. Sudden pituitary apoplexy and blindness have been reported following administration of GnRH to a patient with a gonadotropin-secreting pituitary tumor. Continuous treatment of women with a GnRH analog (leuprolide, nafarelin, goserelin) causes the typical symptoms of menopause, which include hot flushes, sweats, and headaches. Depression, diminished libido, generalized pain, vaginal dryness, and breast atrophy may also occur. Ovarian cysts may develop within the first month of therapy due to its flare effect on gonadotropin secretion and generally resolve after an additional 6 weeks. Reduced bone mineral density and osteoporosis may occur with prolonged use, so patients should be monitored with bone densitometry before repeated treatment courses. Depending on the condition being treated with the GnRH agonist, it may be possible to ameliorate the signs and symptoms of the hypoestrogenic state without losing clinical efficacy by adding back a small dose of a progestin alone or in combination with a low dose of an estrogen. Contraindications to the use of GnRH agonists in women include pregnancy and breast-feeding. In men treated with continuous GnRH agonist administration, adverse effects include hot flushes and sweats, edema, gynecomastia,


decreased libido, decreased hematocrit, reduced bone density, asthenia, and injection site reactions. GnRH analog treatment of children is generally well tolerated. However, temporary exacerbation of precocious puberty may occur during the first few weeks of therapy. Nafarelin nasal spray may cause or aggravate sinusitis.

GNRH RECEPTOR ANTAGONISTS Four synthetic decapeptides that function as competitive antagonists of GnRH receptors are available for clinical use. Ganirelix, cetrorelix, abarelix, and degarelix inhibit the secretion of FSH and LH in a dose-dependent manner. Ganirelix and cetrorelix are approved for use in controlled ovarian stimulation procedures, whereas degarelix and abarelix are approved for men with advanced prostate cancer.

Pharmacokinetics Ganirelix and cetrorelix are absorbed rapidly after subcutaneous injection. Administration of 0.25 mg daily maintains GnRH antagonism. Alternatively, a single 3.0-mg dose of cetrorelix suppresses LH secretion for 96 hours. Degarelix therapy is initiated with 240 mg administered as two subcutaneous injections. Maintenance dosing is with an 80-mg subcutaneous injection every 28 days. The recommended dosage of abarelix is 100 mg administered intramuscularly every 2 weeks for three doses and every 4 weeks thereafter.

Clinical Pharmacology A. Suppression of Gonadotropin Production GnRH antagonists are approved for preventing the LH surge during controlled ovarian stimulation. They offer several advantages over continuous treatment with a GnRH agonist. Because GnRH antagonists produce an immediate antagonist effect, their use can be delayed until day 6–8 of the in vitro fertilization cycle (Figure 37–3), and thus the duration of administration is shorter. They also appear to have a less suppressive effect on the ovarian response to gonadotropin stimulation, which permits a decrease in the total duration and dose of gonadotropin. On the other hand, because their antagonist effects reverse more quickly after their discontinuation, adherence to the treatment regimen is critical. The antagonists produce a more complete suppression of LH secretion than agonists. The suppression of LH may impair follicular development when recombinant or the purified form of FSH is used during an in vitro fertilization cycle. Clinical trials have shown a slightly lower rate of pregnancy in in vitro fertilization cycles that used GnRH antagonist treatment compared with cycles that used GnRH agonist treatment. B. Advanced Prostate Cancer Degarelix and abarelix are approved for the treatment of symptomatic advanced prostate cancer. These GnRH antagonists reduce concentrations of gonadotropins and androgens more rapidly than GnRH agonists and avoid the testosterone surge seen with GnRH agonist therapy.

Toxicity When used for controlled ovarian stimulation, ganirelix and cetrorelix are well tolerated. The most common adverse effects are nausea and headache. During the treatment of men with prostate cancer, degarelix caused injection-site reactions and increases in liver enzymes. Like continuous treatment with a GnRH agonist, degarelix and abarelix lead to signs and symptoms of androgen deprivation, including hot flushes and weight gain.

PROLACTIN Prolactin is a 198-amino-acid peptide hormone produced in the anterior pituitary. Its structure resembles that of GH. Prolactin is the principal hormone responsible for lactation. Milk production is stimulated by prolactin when appropriate circulating levels of estrogens, progestins, corticosteroids, and insulin are present. A deficiency of prolactin—which can occur in rare states of pituitary deficiency—is manifested by failure to lactate or by a luteal phase defect. No preparation of prolactin is available for use in prolactin-deficient patients. In pituitary stalk section from surgery or head trauma, stalk compression due to a sellar mass, or rare cases of hypothalamic destruction, prolactin levels may be elevated as a result of impaired transport of dopamine (prolactin-inhibiting hormone) to the pituitary. Much more commonly, prolactin is elevated as a result of prolactin-secreting adenomas. In addition, a number of drugs elevate prolactin levels. These include antipsychotic and gastrointestinal motility drugs that are known dopamine receptor antagonists, estrogens, and opiates. Hyperprolactinemia causes hypogonadism, which manifests with infertility, oligomenorrhea or amenorrhea, and galactorrhea in premenopausal women, and with loss of libido, erectile dysfunction, and infertility in men. In the case of large tumors (macroadenomas), it can be associated with symptoms of a pituitary mass, including visual changes due to compression of the optic nerves. The


hypogonadism and infertility associated with hyperprolactinemia result from inhibition of GnRH release. For patients with symptomatic hyperprolactinemia, inhibition of prolactin secretion can be achieved with dopamine agonists, which act in the pituitary to inhibit prolactin release.

DOPAMINE AGONISTS Adenomas that secrete excess prolactin usually retain the sensitivity to inhibition by dopamine exhibited by the normal pituitary. Bromocriptine and cabergoline are ergot derivatives (see Chapters 16 and 28) with a high affinity for dopamine D2 receptors. Quinagolide, a drug approved in Europe, is a nonergot agent with similarly high D2 receptor affinity. The chemical structure and pharmacokinetic features of ergot alkaloids are presented in Chapter 16. Dopamine agonists suppress prolactin release very effectively in patients with hyperprolactinemia and GH release is reduced in patients with acromegaly, although not as effectively. Bromocriptine has also been used in Parkinson’s disease to improve motor function and reduce levodopa requirements (see Chapter 28). Newer, nonergot D 2 agonists used in Parkinson’s disease (pramipexole and ropinirole; see Chapter 28) have been reported to interfere with lactation, but they are not approved for use in hyperprolactinemia.

Pharmacokinetics All available dopamine agonists are active as oral preparations, and all are eliminated by metabolism. They can also be absorbed systemically after vaginal insertion of tablets. Cabergoline, with a half-life of approximately 65 hours, has the longest duration of action. Quinagolide has a half-life of about 20 hours, whereas the half-life of bromocriptine is about 7 hours. After vaginal administration, serum levels peak more slowly.

Clinical Pharmacology A. Hyperprolactinemia A dopamine agonist is the standard first-line treatment for hyperprolactinemia. These drugs shrink pituitary prolactin-secreting tumors, lower circulating prolactin levels, and restore ovulation in approximately 70% of women with microadenomas and 30% of women with macroadenomas (Figure 37–4). Cabergoline is initiated at 0.25 mg twice weekly orally or vaginally. It can be increased gradually, according to serum prolactin determinations, up to a maximum of 1 mg twice weekly. Bromocriptine is generally taken daily after the evening meal at the initial dose of 1.25 mg; the dose is then increased as tolerated. Most patients require 2.5–7.5 mg daily. Long-acting oral bromocriptine formulations (Parlodel SRO) and intramuscular formulations (Parlodel L.A.R.) are available outside the United States.


FIGURE 37–4 Results from a clinical trial of cabergoline in women with hyperprolactinemia and anovulation. A: The dashed line indicates the upper limit of normal serum prolactin concentrations. B: Complete success was defined as pregnancy or at least two consecutive menses with evidence of ovulation at least once. Partial success was two menstrual cycles without evidence of ovulation or just one ovulatory cycle. The most common reasons for withdrawal from the trial were nausea, headache, dizziness, abdominal pain, and fatigue. (Adapted from Webster J et al: A comparison of cabergoline and bromocriptine in the treatment of hyperprolactinemic amenorrhea. N Engl J Med 1994;331:904.) B. Physiologic Lactation Dopamine agonists were used in the past to prevent breast engorgement when breast-feeding was not desired. Their use for this purpose has been discouraged because of toxicity (see Toxicity & Contraindications). C. Acromegaly A dopamine agonist alone or in combination with pituitary surgery, radiation therapy, or octreotide administration can be used to treat acromegaly. The doses required are higher than those used to treat hyperprolactinemia. For example, patients with acromegaly require 20–30 mg/d of bromocriptine and seldom respond adequately to bromocriptine alone unless the pituitary tumor secretes prolactin as well as GH.

Toxicity & Contraindications Dopamine agonists can cause nausea, headache, light-headedness, orthostatic hypotension, and fatigue. Psychiatric manifestations occasionally occur, even at lower doses, and may take months to resolve. Erythromelalgia occurs rarely. High dosages of ergot-derived preparations can cause cold-induced peripheral digital vasospasm. Pulmonary infiltrates have occurred with chronic high-dosage therapy.


Cabergoline treatment at high doses for Parkinson’s disease is associated with higher risk of valvular heart disease, but probably not at the lower dose used for hyperprolactinemia. Cabergoline appears to cause nausea less often than bromocriptine. Vaginal administration can reduce nausea, but may cause local irritation. Dopamine agonist therapy during the early weeks of pregnancy has not been associated with an increased risk of spontaneous abortion or congenital malformations. Although there has been a longer experience with the safety of bromocriptine during early pregnancy, there is growing evidence that cabergoline is also safe in women with macroadenomas who must continue a dopamine agonist during pregnancy. In patients with small pituitary adenomas, dopamine agonist therapy is discontinued upon conception because growth of microadenomas during pregnancy is rare. Patients with very large adenomas require vigilance for tumor progression and often require a dopamine agonist throughout pregnancy. There have been rare reports of stroke or coronary thrombosis in postpartum women taking bromocriptine to suppress postpartum lactation.

POSTERIOR PITUITARY HORMONES The two posterior pituitary hormones—vasopressin and oxytocin—are synthesized in neuronal cell bodies in the hypothalamus and transported via their axons to the posterior pituitary, where they are stored and then released into the circulation. Each has limited but important clinical uses.

OXYTOCIN Oxytocin is a peptide hormone secreted by the posterior pituitary. Oxytocin stimulates muscular contractions in the uterus and myoepithelial contractions in the breast. Thus, it is involved in parturition and the letdown of milk. During the second half of pregnancy, uterine smooth muscle shows an increase in the expression of oxytocin receptors and becomes increasingly sensitive to the stimulant action of endogenous oxytocin.

Chemistry & Pharmacokinetics A. Structure Oxytocin is a 9-amino-acid peptide with an intrapeptide disulfide cross-link (Figure 37–5). Its amino acid sequence differs from that of vasopressin at positions 3 and 8.

FIGURE 37–5 Posterior pituitary hormones and desmopressin. (Adapted, with permission, from Ganong WF: Review of Medical Physiology, 21st ed. McGraw-Hill, 2003. Copyright © The McGraw-Hill Companies, Inc.)


B. Absorption, Metabolism, and Excretion Oxytocin is administered intravenously for initiation and augmentation of labor. It also can be administered intramuscularly for control of postpartum bleeding. Oxytocin is not bound to plasma proteins and is rapidly eliminated by the kidneys and liver, with a circulating halflife of 5 minutes.

Pharmacodynamics Oxytocin acts through G protein-coupled receptors and the phosphoinositide-calcium second-messenger system to contract uterine smooth muscle. Oxytocin also stimulates the release of prostaglandins and leukotrienes that augment uterine contraction. In small doses oxytocin increases both the frequency and the force of uterine contractions. At higher doses, it produces sustained contraction. Oxytocin also causes contraction of myoepithelial cells surrounding mammary alveoli, which leads to milk letdown. Without oxytocininduced contraction, normal lactation cannot occur. At high concentrations, oxytocin has weak antidiuretic and pressor activity due to activation of vasopressin receptors.

Clinical Pharmacology Oxytocin is used to induce labor for conditions requiring expedited vaginal delivery such as uncontrolled maternal diabetes, worsening preeclampsia, intrauterine infection, or ruptured membranes after 34 gestational weeks. It is also used to augment protracted labor. Oxytocin can also be used in the immediate postpartum period to stop vaginal bleeding due to uterine atony. Before delivery, oxytocin is usually administered intravenously via an infusion pump with appropriate fetal and maternal monitoring. For induction of labor, an initial infusion rate of 0.5–2 mU/min is increased every 30–60 minutes until a physiologic contraction pattern is established. The maximum infusion rate is 20 mU/min. For postpartum uterine bleeding, 10–40 units are added to 1 L of 5% dextrose, and the infusion rate is titrated to control uterine atony. Alternatively, 10 units of oxytocin can be administered by intramuscular injection. During the antepartum period, oxytocin induces uterine contractions that transiently reduce placental blood flow to the fetus. The oxytocin challenge test measures the fetal heart rate response to a standardized oxytocin infusion and provides information about placental circulatory reserve. An abnormal response, seen as late decelerations in the fetal heart rate, indicates fetal hypoxia and may warrant immediate cesarean delivery.

Toxicity & Contraindications When oxytocin is used judiciously, serious toxicity is rare. The toxicity that does occur is due either to excessive stimulation of uterine contractions or to inadvertent activation of vasopressin receptors. Excessive stimulation of uterine contractions before delivery can cause fetal distress, placental abruption, or uterine rupture. These complications can be detected early by means of standard fetal monitoring. High concentrations of oxytocin with activation of vasopressin receptors can cause excessive fluid retention, or water intoxication, leading to hyponatremia, heart failure, seizures, and death. Bolus injections of oxytocin can cause hypotension. To avoid hypotension, oxytocin is administered intravenously as dilute solutions at a controlled rate. Contraindications to oxytocin include fetal distress, fetal malpresentation, placental abruption, and other predispositions for uterine rupture, including previous extensive uterine surgery.

OXYTOCIN ANTAGONIST Atosiban is an antagonist of the oxytocin receptor that has been approved outside the United States as a treatment (tocolysis) for preterm labor. Atosiban is a modified form of oxytocin that is administered by intravenous infusion for 2–48 hours. In a small number of published clinical trials, atosiban appears to be as effective as β-adrenoceptor-agonist tocolytics and to produce fewer adverse effects. In 1998, however, the FDA decided not to approve atosiban based on concerns about efficacy and safety.

VASOPRESSIN (ANTIDIURETIC HORMONE, ADH) Vasopressin is a peptide hormone released by the posterior pituitary in response to rising plasma tonicity or falling blood pressure. It possesses antidiuretic and vasopressor properties. A deficiency of this hormone results in diabetes insipidus (see also Chapters 15 and 17).

Chemistry & Pharmacokinetics A. Structure


Vasopressin is a nonapeptide with a 6-amino-acid ring and a 3-amino-acid side chain. The residue at position 8 is arginine in humans and in most other mammals except pigs and related species, whose vasopressin contains lysine at position 8 (Figure 37–5). Desmopressin acetate (DDAVP, 1-desamino-8- D-arginine vasopressin) is a long-acting synthetic analog of vasopressin with minimal pressor activity and an antidiuretic-to-pressor ratio 4000 times that of vasopressin. Desmopressin is modified at position 1 and contains a D-amino acid at position 8. Like vasopressin and oxytocin, desmopressin has a disulfide linkage between positions 1 and 6. B. Absorption, Metabolism, and Excretion Vasopressin is administered by intravenous or intramuscular injection. The half-life of circulating vasopressin is approximately 15 minutes, with renal and hepatic metabolism via reduction of the disulfide bond and peptide cleavage. Desmopressin can be administered intravenously, subcutaneously, intranasally, or orally. The half-life of circulating desmopressin is 1.5–2.5 hours. Nasal desmopressin is available as a unit dose spray that delivers 10 mcg per spray; it is also available with a calibrated nasal tube that can be used to deliver a more precise dose. Nasal bioavailability of desmopressin is 3–4%, whereas oral bioavailability is less than 1%.

Pharmacodynamics Vasopressin activates two subtypes of G protein-coupled receptors (see Chapter 17). V1 receptors are found on vascular smooth muscle cells and mediate vasoconstriction via the coupling protein Gq and phospholipase C. V2 receptors are found on renal tubule cells and reduce diuresis through increased water permeability and water resorption in the collecting tubules via Gs and adenylyl cyclase. Extrarenal V2 -like receptors regulate the release of coagulation factor VIII and von Willebrand factor, which increases platelet aggregation.

Clinical Pharmacology Vasopressin and desmopressin are treatments of choice for pituitary diabetes insipidus. The dosage of desmopressin is 10–40 mcg (0.1– 0.4 mL) in two to three divided doses as a nasal spray or, as an oral tablet, 0.1–0.2 mg two to three times daily. The dosage by injection is 1–4 mcg (0.25–1 mL) every 12–24 hours as needed for polyuria, polydipsia, or hypernatremia. Bedtime desmopressin therapy, by intranasal or oral administration, ameliorates nocturnal enuresis by decreasing nocturnal urine production. Vasopressin infusion is effective in some cases of esophageal variceal bleeding and colonic diverticular bleeding. High-dose vasopressin as a 40-unit intravenous bolus injection may be given to replace epinephrine in the Advanced Cardiovascular Life Support (ACLS) resuscitation protocol for pulseless arrest. Desmopressin is also used for the treatment of coagulopathy in hemophilia A and von Willebrand disease (see Chapter 34).

Toxicity & Contraindications Headache, nausea, abdominal cramps, agitation, and allergic reactions occur rarely. Overdosage can result in hyponatremia and seizures. Vasopressin (but not desmopressin) can cause vasoconstriction and should be used cautiously in patients with coronary artery disease. Nasal insufflation of desmopressin may be less effective when nasal congestion is present.

VASOPRESSIN ANTAGONISTS A group of nonpeptide antagonists of vasopressin receptors has been investigated for use in patients with hyponatremia or acute heart failure, which is often associated with elevated concentrations of vasopressin. Conivaptan has high affinity for both V1a and V2 receptors. Tolvaptan has a 30-fold higher affinity for V2 than for V1 receptors. In several clinical trials, both agents promoted the excretion of free water, relieved symptoms, and reduced objective signs of hyponatremia and heart failure. Conivaptan, administered intravenously, and tolvaptan, given orally, are approved by the FDA for treatment of hyponatremia. Tolvaptan treatment duration is limited to 30 days due to risk of hepatotoxicity, including life-threatening liver failure. Several other nonselective nonpeptide vasopressin receptor antagonists are being investigated for these conditions (see Chapter 15).

SUMMARY Hypothalamic & Pituitary Hormones1





PREPARATIONS AVAILABLE



REFERENCES Abramovici A, Cantu J, Jenkins SM: T ocolytic therapy for acute preterm labor. Obstet Gynecol Clin North Am 2012;39:77. Al-Inany HG et al: Gonadotrophin-releasing hormone antagonists for assisted reproductive technology. Cochrane Database Syst Rev 2011;(5):CD001750. Beall SA, DeCherney A: History and challenges surrounding ovarian stimulation in the treatment of infertility. Fertil Steril 2012;97:795. Carel JC et al: Consensus statement on the use of gonadotropin-releasing hormone analogs in children. Pediatrics 2009;123:752. Carel JC et al: Long-term mortality after recombinant growth hormone treatment for isolated growth hormone deficiency or childhood short stature: Preliminary report of the French SAGhE study. J Clin Endo Metab 2012;97:416. Carter-Su C, Schwartz J, Smit LS: Molecular mechanism of growth hormone action. Annu Rev Physiol 1996;58:187. Collett-Solberg PF et al: T he role of recombinant human insulin-like growth factor-I in treating children with short stature. J Clin Endocrinol Metab 2008;93(1):10. Dong Q, Rosenthal SM: Endocrine disorders of the hypothalamus and pituitary. In: Swaiman KF, Ashwal S, Ferriero DM (editors): Pediatric Neurology, 5th ed. Mosby, Inc. (Elsevier, Inc.) 2011. Dreicer R et al: New data, new paradigms for treating prostate cancer patients—VI: Novel hormonal therapy approaches. Urology 2011;78(5 Suppl):S494. Gabe SG et al: Obstetrics: Normal and Problem Pregnancies, 6th ed. Churchill Livingstone, 2012. Han T S, Bouloux PM: What is the optimal therapy for young males with hypogonadatropic hypogonadism? Clin Endocrinol 2010;72:731. Hodson EM, Willis NS, Craig JC: Growth hormone for children with chronic kidney disease. Cochrane Database Syst Rev 2012;(2):CD003264. Katznelson L et al: American Association of Clinical Endocrinologists medical guidelines for clinical practice for the diagnosis and treatment of acromegaly—2011 update. Endo Pract 2011;s4:1. Mammen AA, Ferrer FA: Nocturnal enuresis: Medical management. Urol Clin North Am 2004;31:491. Melmed S et al (editors): Williams Textbook of Endocrinology, 12th ed. Saunders, 2011. Papatsonis DN et al: Maintenance therapy with oxytocin antagonists for inhibiting preterm birth after threatened preterm labour. Cochrane Database Syst Rev 2013; (10):CD005938. Penson D et al: Effectiveness of hormonal and surgical therapies for cryptorchidism: A systematic review. Pediatrics 2013;131:e1897. Richmond E, Rogol AD: Current indications for growth hormone therapy for children and adolescents. Endocr Dev 2010;18:92. Rosenfeld RG, Hwa V: T he growth hormone cascade and its role in mammalian growth. Horm Res 2009;71(Suppl 2):36. Sävendahl L et al: Long-term mortality and causes of death in isolated GHD, ISS, and SGA patients treated with recombinant growth hormone during childhood in Belgium, T he Netherlands, and Sweden: Preliminary report of 3 countries participating in the EU SAGhE study. J Clin Endo Metab 2012;97:E213. Speroff L, Fritz MA: Clinical Gynecologic Endocrinology and Infertility, 8th ed. Lippincott Williams & Wilkins, 2010. Strauss JF, Barbieri RL: Yen & Jaffe’s Reproductive Endocrinology, 6th ed. Elsevier, 2009. Surrey ES: Gonadotropin-releasing hormone agonist and add-back therapy: What do the data show? Curr Opin Obstet Gynecol 2010;22:283. Synder PJ: T reatment of hyperprolactinemia due to lactotroph adenoma and other causes. www.UpT oDate.com. T akala J et al: Increased mortality associated with growth hormone treatment in critically ill adults. N Engl J Med 1999;341:785. T ena-Sempere M: Deciphering puberty: Novel partners, novel mechanisms. Eur J Endocrinol 2012;167:733. Wales PW et al: Human growth hormone and glutamine for patients with short bowel syndrome. Cochrane Database Syst Rev 2010;(16):CD006321. Webster J et al: A comparison of cabergoline and bromocriptine in the treatment of hyperprolactinemic amenorrhea. N Engl J Med 1994;331:904. Westhoff G, Cotter AM, T olosa JE: Prophylactic oxytocin for the third stage of labour to prevent postpartum haemorrhage. Cochrane Database Syst Rev 2013; (10):CD001808. Wit JM et al: Idiopathic short stature: Definition, epidemiology, and diagnostic evaluation. Growth Horm IGF Res 2008;18:89. Youssef MA et al: Gonadotropin-releasing hormone agonist versus HCG for oocyte triggering in antagonist assisted reproductive technology cycles. Cochrane Database Syst Rev 2011;(1):CD008046.

CASE STUDY ANSWER While growth hormone (GH) may have some direct growth-promoting effects, it is thought to mediate skeletal growth principally through epiphyseal production of insulin-like growth factor-I (IGF-I), which acts mainly in an autocrine/paracrine manner. IGF-I may also promote statural growth through endocrine mechanisms. The findings of small testes and a microphallus in this patient suggest a diagnosis of hypogonadism, likely as a consequence of gonadotropin deficiency. This patient is at risk for multiple hypothalamic/pituitary deficiencies. He may already have or may subsequently develop ACTH/cortisol and TSH/thyroid hormone deficiencies and thus may require supplementation with hydrocortisone and levothyroxine, in addition to supplementation with GH and testosterone. He should also be evaluated for the presence of central diabetes insipidus and, if present, treated with desmopressin, a V2 vasopressin receptor-selective analog.


CHAPTER

38 Thyroid & Antithyroid Drugs Betty J. Dong, PharmD, FASHP, FCCP, & Francis S. Greenspan, MD, FACP

CASE STUDY A 33-year-old woman presents with complaints of fatigue, sluggishness, weight gain, cold intolerance, dry skin, and muscle weakness for the last 2 months. She is so tired that she has to take several naps during the day to complete her tasks. These complaints are new for her since she used to feel warm all the time, had boundless energy causing her some insomnia, and states she felt like her heart was going to jump out of her chest. She also states that she would like to become pregnant in the near future. Her past medical history is significant for radioactive iodine therapy (RAI) about 1 year ago after a short trial of methimazole and propranolol therapy. She underwent RAI due to her poor medication adherence and did not attend routine scheduled appointments afterward. On physical examination, her blood pressure is 130/89 mm Hg with a pulse of 50 bpm. Her weight is 136 lb (61.8 kg), an increase of 10 lb (4.5 kg) in the last year. Her thyroid gland is not palpable and her reflexes are delayed. Laboratory findings include a thyroid-stimulating hormone (TSH) level of 24.9 μIU/mL and a free thyroxine level of 8 pmol/L. Evaluate the management of her past history of hyperthyroidism. Identify the available treatment options for control of her current thyroid status.

THYROID PHYSIOLOGY The normal thyroid gland secretes sufficient amounts of the thyroid hormones—triiodothyronine (T3 ) and tetraiodothyronine (T4 , thyroxine)—to normalize growth and development, body temperature, and energy levels. These hormones contain 59% and 65% (respectively) of iodine as an essential part of the molecule. Calcitonin, the second type of thyroid hormone, is important in the regulation of calcium metabolism and is discussed in Chapter 42.

Iodide Metabolism The recommended daily adult iodide (I−)* intake is 150 mcg (200 mcg during pregnancy and lactation). Iodide, ingested from food, water, or medication, is rapidly absorbed and enters an extracellular fluid pool. The thyroid gland removes about 75 mcg a day from this pool for hormone synthesis, and the balance is excreted in the urine. If iodide intake is increased, the fractional iodine uptake by the thyroid is diminished.

Biosynthesis of Thyroid Hormones Once taken up by the thyroid gland, iodide undergoes a series of enzymatic reactions that incorporate it into active thyroid hormone (Figure 38–1). The first step is the transport of iodide into the thyroid gland by an intrinsic follicle cell basement membrane protein called the sodium/iodide symporter (NIS). This can be inhibited by such anions as thiocyanate (SCN−), pertechnetate (TcO 4 −), and perchlorate (CIO4 −). At the apical cell membrane a second I − transport enzyme called pendrin controls the flow of iodide across the membrane. Pendrin is also found in the cochlea of the inner ear. If pendrin is deficient or absent (PDS or SLC26A4 mutation), a hereditary syndrome of goiter and deafness, called Pendred’s syndrome, ensues. At the apical cell membrane, iodide is oxidized by thyroidal peroxidase (TPO) to iodine, in which form it rapidly iodinates tyrosine residues within the thyroglobulin molecule to form monoiodotyrosine (MIT) and diiodotyrosine (DIT). This process is called iodide organification. Thyroidal peroxidase is transiently blocked by high levels of intrathyroidal iodide and blocked more persistently by thioamide drugs. Gene expression of TPO is stimulated by thyroid-stimulating hormone (TSH).


FIGURE 38–1 Biosynthesis of thyroid hormones. The sites of action of various drugs that interfere with thyroid hormone biosynthesis are shown. Two molecules of DIT combine within the thyroglobulin molecule to form L-thyroxine (T4 ). One molecule of MIT and one molecule of DIT combine to form T3 . In addition to thyroglobulin, other proteins within the gland may be iodinated, but these iodoproteins do not have hormonal activity. Thyroxine, T3 , MIT, and DIT are released from thyroglobulin by exocytosis and proteolysis of thyroglobulin at the apical colloid border. The MIT and DIT are then deiodinated within the gland, and the iodine is reutilized. This process of proteolysis is also blocked by high levels of intrathyroidal iodide. The ratio of T4 to T3 within thyroglobulin is approximately 5:1, so that most of the hormone released is thyroxine. Most of the T3 circulating in the blood is derived from peripheral metabolism of thyroxine (see below, Figure 38–2).


FIGURE 38–2 Peripheral metabolism of thyroxine. (Adapted, with permission, from Gardner DG, Shoback D [editors]: Greenspan’s Basic & Clinical Endocrinology, 8th ed. McGraw-Hill, 2007. Copyright © The McGraw-Hill Companies, Inc.)

Transport of Thyroid Hormones Thyroxine and T3 in plasma are reversibly bound to protein, primarily thyroxine-binding globulin (TBG). Only about 0.04% of total T4 and 0.4% of T3 exist in the free form (as FT4 and FT3 ). Many physiologic and pathologic states and drugs affect T4 , T3 , and thyroid transport. However, the actual levels of free hormone generally remain normal, reflecting feedback control.

Peripheral Metabolism of Thyroid Hormones The primary pathway for the peripheral metabolism of thyroxine is deiodination by three 5′deiodinase enzymes (D1, D2, D3). Deiodination of T4 may occur by monodeiodination of the outer ring, producing 3,5,3′-triiodothyronine (T3 ), which is three to four times more potent than T4 . The D1 enzyme is responsible for most of the circulating T3 while D2 regulates T3 levels in the brain and pituitary. D3 deiodination produces metabolically inactive 3,3′,5′-triiodothyronine (reverse T3 [rT3 ]), (Figure 38–2). The low serum levels of T3 and rT3 in normal individuals are due to the high metabolic clearances of these two compounds. Drugs such as amiodarone, iodinated contrast media, β blockers, and corticosteroids, and severe illness or starvation inhibit the 5′deiodinase necessary for the conversion of T4 to T3 , resulting in low T3 and high rT3 levels in the serum. A polymorphism in the D2 gene can reduce T3 activation and impair thyroid hormone response. The pharmacokinetics of thyroid hormones are listed in Table 38–1. TABLE 38–1 Summary of thyroid hormone kinetics.


Evaluation of Thyroid Function The tests used to evaluate thyroid function are listed in Table 38–2. TABLE 38–2 Typical values for thyroid function tests.


A. Thyroid-Pituitary Relationships Control of thyroid function via thyroid-pituitary feedback is also discussed in Chapter 37. Hypothalamic cells secrete thyrotropin-releasing hormone (TRH) (Figure 38–3). TRH is secreted into capillaries of the pituitary portal venous system, and in the pituitary gland, TRH stimulates the synthesis and release of thyrotropin (thyroid-stimulating hormone, TSH). TSH in turn stimulates an adenylyl cyclase– mediated mechanism in the thyroid cell to increase the synthesis and release of T4 and T3 . These thyroid hormones act in a negative feedback fashion in the pituitary to block the action of TRH and in the hypothalamus to inhibit the synthesis and secretion of TRH. Other hormones or drugs may also affect the release of TRH or TSH.


FIGURE 38–3 The hypothalamic-pituitary-thyroid axis. Acute psychosis or prolonged exposure to cold may activate the axis. Hypothalamic thyroid-releasing hormone (TRH) stimulates pituitary thyroid-stimulating hormone (TSH) release, while somatostatin and dopamine inhibit it. TSH stimulates T4 and T3 synthesis and release from the thyroid, and they in turn inhibit both TRH and TSH synthesis and release. Small amounts of iodide are necessary for hormone production, but large amounts inhibit T3 and T4 production and release. Solid arrows, stimulatory influence; dashed arrows, inhibitory influence. H, hypothalamus; AP, anterior pituitary. B. Autoregulation of the Thyroid Gland The thyroid gland also regulates its uptake of iodide and thyroid hormone synthesis by intrathyroidal mechanisms that are independent of TSH. These mechanisms are primarily related to the level of iodine in the blood. Large doses of iodine inhibit iodide organification (Wolff-Chaikoff block, see Figure 38–1). In certain disease states (eg, Hashimoto’s thyroiditis), this can inhibit thyroid hormone synthesis and result in hypothyroidism. Hyperthyroidism can result from the loss of the Wolff-Chaikoff block in susceptible individuals (eg, multinodular goiter). C. Abnormal Thyroid Stimulators In Graves’ disease (see below), lymphocytes secrete a TSH receptor-stimulating antibody (TSH-R Ab [stim]), also known as thyroidstimulating immunoglobulin (TSI). This immunoglobulin binds to the TSH receptor and stimulates the gland in the same fashion as TSH itself. The duration of its effect, however, is much longer than that of TSH. TSH receptors are also found in orbital fibrocytes, which


may be stimulated by high levels of TSH-R Ab [stim] and can cause ophthalmopathy.

BASIC PHARMACOLOGY OF THYROID & ANTITHYROID DRUGS THYROID HORMONES Chemistry The structural formulas of thyroxine and triiodothyronine as well as reverse triiodothyronine (rT3 ) are shown in Figure 38–2. All of these naturally occurring molecules are levo (L) isomers. The synthetic dextro (D) isomer of thyroxine, dextrothyroxine, has approximately 4% of the biologic activity of the L-isomer as evidenced by its lesser ability to suppress TSH secretion and correct hypothyroidism.

Pharmacokinetics Thyroxine is absorbed best in the duodenum and ileum; absorption is modified by intraluminal factors such as food, drugs, gastric acidity, and intestinal flora. Oral bioavailability of current preparations of L-thyroxine averages 70% (Table 38–1). In contrast, T3 is almost completely absorbed (95%). T4 and T3 absorption appears not to be affected by mild hypothyroidism but may be impaired in severe myxedema with ileus. These factors are important in switching from oral to parenteral therapy. For parenteral use, the intravenous route is preferred for both hormones. In patients with hyperthyroidism, the metabolic clearances of T4 and T3 are increased and the half-lives decreased; the opposite is true in patients with hypothyroidism. Drugs that induce hepatic microsomal enzymes (eg, rifampin, phenobarbital, carbamazepine, phenytoin, tyrosine kinase inhibitors, HIV protease inhibitors) increase the metabolism of both T4 and T3 (Table 38–3). Despite this change in clearance, the normal hormone concentration is maintained in the majority of euthyroid patients as a result of compensatory hyperfunction of the thyroid. However, patients dependent on T 4 replacement medication may require increased dosages to maintain clinical effectiveness. A similar compensation occurs if binding sites are altered. If TBG sites are increased by pregnancy, estrogens, or oral contraceptives, there is an initial shift of hormone from the free to the bound state and a decrease in its rate of elimination until the normal free hormone concentration is restored. Thus, the concentration of total and bound hormone will increase, but the concentration of free hormone and the steady-state elimination will remain normal. The reverse occurs when thyroid binding sites are decreased. TABLE 38–3 Drug effects and thyroid function.



Mechanism of Action A model of thyroid hormone action is depicted in Figure 38–4, which shows the free forms of thyroid hormones, T4 and T3 , dissociated from thyroid-binding proteins, entering the cell by the active transporters (eg, monocarboxylate transporter 8 [MCT8], MCT10, and organic anion transporting polypeptide [OATP1C1]). Transporter mutations can result in a clinical syndrome of mental retardation, myopathy, and low serum T 4 levels (Allan-Herndon-Dudley syndrome). Within the cell, T 4 is converted to T3 by 5′-deiodinase, and the T3 enters the nucleus, where T3 binds to a specific T3 thyroid receptor protein, a member of the c-erb oncogene family. (This family also includes the steroid hormone receptors and receptors for vitamins A and D.) The T 3 receptor exists in two forms, α and β. Mutations in both α and β genes have been associated with generalized thyroid hormone resistance. Differing concentrations of receptor forms in different tissues may account for variations in T3 effect on these tissues.


FIGURE 38–4 Model of the interaction of T3 with the T3 receptor. A: Inactive phase—the unliganded T3 receptor dimer bound to the thyroid hormone response element (TRE) along with corepressors acts as a suppressor of gene transcription. B: Active phase—T3 and T4 circulate bound to thyroid-binding proteins (TBPs). The free hormones are transported into the cell by a specific transport system. Within the cytoplasm, T4 is converted to T3 by 5′-deiodinase (5′DI); T3 then moves into the nucleus. There it binds to the ligand-binding domain of the thyroid receptor (TR) monomer. This promotes disruption of the TR homodimer and heterodimerization with retinoid X


receptor (RXR) on the TRE, displacement of corepressors, and binding of coactivators. The TR-coactivator complex activates gene transcription, which leads to alteration in protein synthesis and cellular phenotype. TR-LBD, T3 receptor ligand-binding domain; TRDBD, T3 receptor DNA-binding domain; RXR-LBD, retinoid X receptor ligand-binding domain; RXR-DBD, retinoid X receptor DNAbinding domain; T3 , triiodothyronine; T4 , tetraiodothyronine, L-thyroxine. (Adapted, with permission, from Gardner DG, Shoback D [editors]: Greenspan’s Basic & Clinical Endocrinology, 8th ed. McGraw-Hill, 2007. Copyright © The McGraw-Hill Companies, Inc.) Most of the effects of thyroid on metabolic processes appear to be mediated by activation of nuclear receptors that lead to increased formation of RNA and subsequent protein synthesis, eg, increased formation of Na +/K+-ATPase. This is consistent with the observation that the action of thyroid is manifested in vivo with a time lag of hours or days after its administration. Large numbers of thyroid hormone receptors are found in the most hormone-responsive tissues (pituitary, liver, kidney, heart, skeletal muscle, lung, and intestine), while few receptor sites occur in hormone-unresponsive tissues (spleen, testes). The brain, which lacks an anabolic response to T3 , contains an intermediate number of receptors. In congruence with their biologic potencies, the affinity of the receptor site for T4 is about ten times lower than that for T3 . Under some conditions, the number of nuclear receptors may be altered to preserve body homeostasis. For example, starvation lowers both circulating T3 hormone and cellular T3 receptors.

Effects of Thyroid Hormones The thyroid hormones are responsible for optimal growth, development, function, and maintenance of all body tissues. Excess or inadequate amounts result in the signs and symptoms of hyperthyroidism or hypothyroidism, respectively (Table 38–4). Since T3 and T4 are qualitatively similar, they may be considered as one hormone in the discussion that follows. TABLE 38–4 Manifestations of thyrotoxicosis and hypothyroidism.



Thyroid hormone is critical for the development and functioning of nervous, skeletal, and reproductive tissues. Its effects depend on protein synthesis as well as potentiation of the secretion and action of growth hormone. Thyroid deprivation in early life results in irreversible mental retardation and dwarfism—typical of congenital cretinism. Effects on growth and calorigenesis are accompanied by a pervasive influence on metabolism of drugs as well as carbohydrates, fats, proteins, and vitamins. Many of these changes are dependent upon or modified by activity of other hormones. Conversely, the secretion and degradation rates of virtually all other hormones, including catecholamines, cortisol, estrogens, testosterone, and insulin, are affected by thyroid status. Many of the manifestations of thyroid hyperactivity resemble sympathetic nervous system overactivity (especially in the cardiovascular system), although catecholamine levels are not increased. Changes in catecholamine-stimulated adenylyl cyclase activity as measured by cAMP are found with changes in thyroid activity. Thyroid hormone increases the numbers of β receptors and enhances amplification of the β-receptor signal. Other clinical symptoms reminiscent of excessive epinephrine activity (and partially alleviated by adrenoceptor antagonists) include lid lag and retraction, tremor, excessive sweating, anxiety, and nervousness. The opposite constellation of effects is seen in hypothyroidism (Table 38–4).

Thyroid Preparations See the Preparations Available section at the end of this chapter for a list of available preparations. These preparations may be synthetic (levothyroxine, liothyronine, liotrix) or of animal origin (desiccated thyroid). Thyroid hormones are not effective and can be detrimental in the management of obesity, abnormal vaginal bleeding, or depression if thyroid hormone levels are normal. Anecdotal reports of a beneficial effect of T 3 administered with antidepressants were not confirmed in a controlled study. Synthetic levothyroxine is the preparation of choice for thyroid replacement and suppression therapy because of its stability, content uniformity, low cost, lack of allergenic foreign protein, easy laboratory measurement of serum levels, and long half-life (7 days), which permits once-daily to weekly administration. In addition, T4 is converted to T3 intracellularly; thus, administration of T4 produces both hormones and T3 administration is unnecessary. Generic levothyroxine preparations provide comparable efficacy and are more costeffective than branded preparations, It is preferable that patients remain on a consistent levothyroxine preparation between refills to avoid changes in bioavailability. A branded soft gel capsule (Tirosint) had faster, more complete dissolution and was less affected by gastric pH or coffee than a tablet formulation. Although liothyronine (T3 ) is three to four times more potent than levothyroxine, it is not recommended for routine replacement therapy because of its shorter half-life (24 hours), requiring multiple daily doses, and difficulty in monitoring its adequacy of replacement by conventional laboratory tests. T3 should also be avoided in patients with cardiac disease due to significant elevations in peak levels and a greater risk of cardiotoxicity. Using the more expensive thyroxine and liothyronine fixed-dose combination (liotrix) and desiccated thyroid has not been shown to be more effective than T4 administration alone. T3 is best reserved for short-term TSH suppression. Research is ongoing to clarify whether T3 might be more appropriate in patients with a polymorphism in the D2 gene who continue to report fatigue, weight gain, and mental impairment while on T4 alone. The use of desiccated thyroid rather than synthetic preparations is never justified, since the disadvantages of protein antigenicity, product instability, variable hormone concentrations, and difficulty in laboratory monitoring far outweigh the advantage of lower cost. Significant amounts of T3 found in some thyroid extracts may produce significant elevations in T3 levels and toxicity. Equi-effective doses are 60 mg of desiccated thyroid, 88–100 mcg of levothyroxine, and approximately 37.5 mcg of liothyronine. The shelf life of synthetic hormone preparations is about 2 years, particularly if they are stored in dark bottles to minimize spontaneous deiodination. The shelf life of desiccated thyroid is not known with certainty, but its potency is better preserved if it is kept dry.

ANTITHYROID AGENTS Reduction of thyroid activity and hormone effects can be accomplished by agents that interfere with the production of thyroid hormones, by agents that modify the tissue response to thyroid hormones, or by glandular destruction with radiation or surgery. Goitrogens are agents that suppress secretion of T3 and T4 to subnormal levels and thereby increase TSH, which in turn produces glandular enlargement (goiter). The antithyroid compounds used clinically include the thioamides, iodides, and radioactive iodine.

THIOAMIDES The thioamides methimazole and propylthiouracil are major drugs for treatment of thyrotoxicosis. In the United Kingdom, carbimazole, which is converted to methimazole in vivo, is widely used. Methimazole is about ten times more potent than propylthiouracil and is the drug of choice in adults and children. Due to a black box warning about severe hepatitis, propylthiouracil should be reserved for use during the first trimester of pregnancy, in thyroid storm, and in those experiencing adverse reactions to methimazole (other than


agranulocytosis or hepatitis). The chemical structures of these compounds are shown in Figure 38–5. The thiocarbamide group is essential for antithyroid activity.

FIGURE 38–5 Structure of thioamides. The thiocarbamide moiety is shaded in color.

Pharmacokinetics Methimazole is completely absorbed but at variable rates. It is readily accumulated by the thyroid gland and has a volume of distribution similar to that of propylthiouracil. Excretion is slower than with propylthiouracil; 65–70% of a dose is recovered in the urine in 48 hours. In contrast, propylthiouracil is rapidly absorbed, reaching peak serum levels after 1 hour. The bioavailability of 50–80% may be due to incomplete absorption or a large first-pass effect in the liver. The volume of distribution approximates total body water with accumulation in the thyroid gland. Most of an ingested dose of propylthiouracil is excreted by the kidney as the inactive glucuronide within 24 hours. The short plasma half-life of these agents (1.5 hours for propylthiouracil and 6 hours for methimazole) has little influence on the duration of the antithyroid action or the dosing interval because both agents are accumulated by the thyroid gland. For propylthiouracil, giving the drug every 6–8 hours is reasonable since a single 100 mg dose can inhibit iodine organification by 60% for 7 hours. Since a single 30 mg dose of methimazole exerts an antithyroid effect for longer than 24 hours, a single daily dose is effective in the management of mild to severe hyperthyroidism. Both thioamides cross the placental barrier and are concentrated by the fetal thyroid, so that caution must be employed when using these drugs in pregnancy. Because of the risk of fetal hypothyroidism, both thioamides are classified as FDA pregnancy category D (evidence of human fetal risk based on adverse reaction data from investigational or marketing experience, see Chapter 59). Of the two, propylthiouracil is preferable during the first trimester of pregnancy because it is more strongly protein-bound and, therefore, crosses the placenta less readily. In addition, methimazole has been, albeit rarely, associated with congenital malformations. Both thioamides are secreted in low concentrations in breast milk but are considered safe for the nursing infant.

Pharmacodynamics The thioamides act by multiple mechanisms. The major action is to prevent hormone synthesis by inhibiting the thyroid peroxidasecatalyzed reactions and blocking iodine organification. In addition, they block coupling of the iodotyrosines. They do not block uptake of iodide by the gland. Propylthiouracil but not methimazole inhibits the peripheral deiodination of T4 and T3 (Figure 38–1). Since the synthesis rather than the release of hormones is affected, the onset of these agents is slow, often requiring 3–4 weeks before stores of


T4 are depleted.

Toxicity Adverse reactions to the thioamides occur in 3–12% of treated patients. Most reactions occur early, especially nausea and gastrointestinal distress. An altered sense of taste or smell may occur with methimazole. The most common adverse effect is a maculopapular pruritic rash (4–6%), at times accompanied by systemic signs such as fever. Rare adverse effects include an urticarial rash, vasculitis, a lupus-like reaction, lymphadenopathy, hypoprothrombinemia, exfoliative dermatitis, polyserositis, and acute arthralgia. An increased risk of severe hepatitis, sometimes resulting in death, has been reported with propylthiouracil (black box warning), so it should be avoided in children and adults unless no other options are available. Cholestatic jaundice is more common with methimazole than propylthiouracil. Asymptomatic elevations in transaminase levels can also occur. The most dangerous complication is agranulocytosis (granulocyte count < 500 cells/mm3 ), an infrequent but potentially fatal adverse reaction. It occurs in 0.1–0.5% of patients taking thioamides, but the risk may be increased in older patients and in those receiving more than 40 mg/d of methimazole. The reaction is usually rapidly reversible when the drug is discontinued, but broad-spectrum antibiotic therapy may be necessary for complicating infections. Colony-stimulating factors (eg, G-CSF; see Chapter 33) may hasten recovery of the granulocytes. The cross-sensitivity between propylthiouracil and methimazole is about 50%; therefore, switching drugs in patients with severe reactions is not recommended.

ANION INHIBITORS Monovalent anions such as perchlorate (ClO4 −), pertechnetate (TcO4 −), and thiocyanate (SCN−) can block uptake of iodide by the gland through competitive inhibition of the iodide transport mechanism. Since these effects can be overcome by large doses of iodides, their effectiveness is somewhat unpredictable. The major clinical use for potassium perchlorate is to block thyroidal reuptake of I− in patients with iodide-induced hyperthyroidism (eg, amiodarone-induced hyperthyroidism). However, potassium perchlorate is rarely used clinically because it is associated with aplastic anemia.

IODIDES Prior to the introduction of the thioamides in the 1940s, iodides were the major antithyroid agents; today they are rarely used as sole therapy.

Pharmacodynamics Iodides have several actions on the thyroid. They inhibit organification and hormone release and decrease the size and vascularity of the hyperplastic gland. In susceptible individuals, iodides can induce hyperthyroidism (Jod-Basedow phenomenon) or precipitate hypothyroidism. In pharmacologic doses (> 6 mg/d), the major action of iodides is to inhibit hormone release, possibly through inhibition of thyroglobulin proteolysis. Improvement in thyrotoxic symptoms occurs rapidly—within 2–7 days—hence the value of iodide therapy in thyroid storm. In addition, iodides decrease the vascularity, size, and fragility of a hyperplastic gland, making the drugs valuable as preoperative preparation for surgery.

Clinical Use of Iodide Disadvantages of iodide therapy include an increase in intraglandular stores of iodine, which may delay onset of thioamide therapy or prevent use of radioactive iodine therapy for several weeks. Thus, iodides should be initiated after onset of thioamide therapy and avoided if treatment with radioactive iodine seems likely. Iodide should not be used alone, because the gland will escape from the iodide block in 2–8 weeks, and its withdrawal may produce severe exacerbation of thyrotoxicosis in an iodine-enriched gland. Chronic use of iodides in pregnancy should be avoided, since they cross the placenta and can cause fetal goiter. In radiation emergencies involving release of radioactive iodine isotopes, the thyroid-blocking effects of potassium iodide can protect the gland from subsequent damage if administered before radiation exposure.

Toxicity Adverse reactions to iodine (iodism) are uncommon and in most cases reversible upon discontinuance. They include acneiform rash


(similar to that of bromism), swollen salivary glands, mucous membrane ulcerations, conjunctivitis, rhinorrhea, drug fever, metallic taste, bleeding disorders, and rarely, anaphylactoid reactions.

RADIOACTIVE IODINE 131

I is the only isotope used for treatment of thyrotoxicosis (others are used in diagnosis). Administered orally in solution as sodium 131 I, it is rapidly absorbed, concentrated by the thyroid, and incorporated into storage follicles. Its therapeutic effect depends on emission of β rays with an effective half-life of 5 days and a penetration range of 400–2000 μm. Within a few weeks after administration, destruction of the thyroid parenchyma is evidenced by epithelial swelling and necrosis, follicular disruption, edema, and leukocyte infiltration. Advantages of radioiodine include easy administration, effectiveness, low expense, and absence of pain. Fears of radiation-induced genetic damage, leukemia, and neoplasia have not been realized after more than 50 years of clinical experience with radioiodine therapy for hyperthyroidism. Radioactive iodine should not be administered to pregnant women or nursing mothers, since it crosses the placenta to destroy the fetal thyroid gland and it is excreted in breast milk.

ADRENOCEPTOR-BLOCKING AGENTS Beta blockers without intrinsic sympathomimetic activity (eg, metoprolol, propranolol, atenolol) are effective therapeutic adjuncts in the management of thyrotoxicosis since many of these symptoms mimic those associated with sympathetic stimulation. Propranolol has been the β blocker most widely studied and used in the therapy of thyrotoxicosis. Beta blockers cause clinical improvement of hyperthyroid symptoms but do not typically alter thyroid hormone levels. Propranolol at doses greater than 160 mg/d may also reduce T3 levels approximately 20% by inhibiting the peripheral conversion of T4 to T3 .

CLINICAL PHARMACOLOGY OF THYROID & ANTITHYROID DRUGS HYPOTHYROIDISM Hypothyroidism is a syndrome resulting from deficiency of thyroid hormones and is manifested largely by a reversible slowing down of all body functions (Table 38–4). In infants and children, there is striking retardation of growth and development that results in dwarfism and irreversible mental retardation. The etiology and pathogenesis of hypothyroidism are outlined in Table 38–5. Hypothyroidism can occur with or without thyroid enlargement (goiter). The laboratory diagnosis of hypothyroidism in the adult is easily made by the combination of low free thyroxine and elevated serum TSH levels (Table 38–2). TABLE 38–5 Etiology and pathogenesis of hypothyroidism.


The most common cause of hypothyroidism in the USA at this time is probably Hashimoto’s thyroiditis, an immunologic disorder in genetically predisposed individuals. In this condition, there is evidence of humoral immunity in the presence of antithyroid antibodies and lymphocyte sensitization to thyroid antigens. Genetic mutations as discussed previously and certain medications can also cause hypothyroidism (Table 38–5).

MANAGEMENT OF HYPOTHYROIDISM Except for hypothyroidism caused by drugs, which can be treated in some cases by simply removing the depressant agent, the general strategy of replacement therapy is appropriate. The most satisfactory preparation is levothyroxine, administered as either a branded or generic preparation. Multiple trials have documented that combination levothyroxine plus liothyronine is not superior to levothyroxine alone. There is some variability in the absorption of thyroxine; dosage will also vary depending on age and weight. Infants and children require more T4 per kilogram of body weight than adults. The average dosage for an infant 1–6 months of age is 10–15 mcg/kg/d, whereas the average dosage for an adult is about 1.7 mcg/kg/d (0.8 mcg/lb/d) or 125 mcg/d. Older adults (> 65 years of age) may require less thyroxine (1.6 mcg/kg/d or 0.7 mcg/lb/d) for replacement. In patients requiring suppression therapy post-thyroidectomy for thyroid cancer, the average dosage of T4 is about 2.2 mcg/kg/d (1 mcg/lb/d). Since interactions with certain foods (eg, bran, soy, coffee) and drugs (Table 38–3) can impair its absorption, thyroxine should be administered on an empty stomach (eg, 60 minutes before meals, 4 hours after meals, or at bedtime) to maintain TSH within an optimal range of 0.5–2.5 mIU/L. Its long half-life of 7 days permits once-daily dosing. Children should be monitored for normal growth and development. Serum TSH and free thyroxine should always be measured before thyroxine administration to avoid transient serum alterations. It takes 6–8 weeks after starting a given dose of thyroxine to reach steady-state levels in the bloodstream. Thus, dosage changes should be made slowly. In younger patients or those with very mild disease, full replacement therapy may be started immediately. In older patients (> 50 years) without cardiac disease, levothyroxine can be started at a dosage of 50 mcg/d. In long-standing hypothyroidism and in older patients with underlying cardiac disease, it is imperative to start with reduced dosages of levothyroxine, 12.5–25 mcg/d for 2 weeks, before increasing by 12.5–25 mcg/d every 2 weeks until euthyroidism or drug toxicity is observed. In cardiac patients, the heart is very sensitive to the level of circulating thyroxine, and if angina pectoris or cardiac arrhythmia develops, it is essential to stop or reduce the thyroxine dosage immediately.


Thyroxine toxicity is directly related to the hormone level. In children, restlessness, insomnia, and accelerated bone maturation and growth may be signs of thyroxine toxicity. In adults, increased nervousness, heat intolerance, episodes of palpitation and tachycardia, or unexplained weight loss may be the presenting symptoms. If these symptoms are present, it is important to monitor serum TSH and FT4 levels (Table 38–2), which will determine whether the symptoms are due to excess thyroxine blood levels. Chronic overtreatment with T4 , particularly in elderly patients, can increase the risk of atrial fibrillation and accelerated osteoporosis.

Special Problems in Management of Hypothyroidism A. Myxedema and Coronary Artery Disease Since myxedema frequently occurs in older persons, it is often associated with underlying coronary artery disease. In this situation, the low levels of circulating thyroid hormone actually protect the heart against increasing demands that could result in angina pectoris, atrial fibrillation, or myocardial infarction. Correction of myxedema must be done cautiously to avoid provoking these cardiac events. If coronary artery surgery is indicated, it should be done first, prior to correction of the myxedema by thyroxine administration. B. Myxedema Coma Myxedema coma is an end state of untreated hypothyroidism. It is associated with progressive weakness, stupor, hypothermia, hypoventilation, hypoglycemia, hyponatremia, water intoxication, shock, and death. Myxedema coma is a medical emergency. The patient should be treated in the intensive care unit, since tracheal intubation and mechanical ventilation may be required. Associated illnesses such as infection or heart failure must be treated by appropriate therapy. It is important to give all preparations intravenously, because patients with myxedema coma absorb drugs poorly from other routes. Intravenous fluids should be administered with caution to avoid excessive water intake. These patients have large pools of empty T3 and T4 binding sites that must be filled before there is adequate free thyroxine to affect tissue metabolism. Accordingly, the treatment of choice in myxedema coma is to give a loading dose of levothyroxine intravenously—usually 300–400 mcg initially, followed by 50–100 mcg daily. Intravenous T 3 can also be used but may be more cardiotoxic and more difficult to monitor. Intravenous hydrocortisone is indicated if the patient has associated adrenal or pituitary insufficiency but is probably not necessary in most patients with primary myxedema. Opioids and sedatives must be used with extreme caution. C. Hypothyroidism and Pregnancy Hypothyroid women frequently have anovulatory cycles and are therefore relatively infertile until restoration of the euthyroid state. This has led to the widespread use of thyroid hormone for infertility, although there is no evidence for its usefulness in infertile euthyroid patients. In a pregnant hypothyroid patient receiving thyroxine, it is extremely important that the daily dose of thyroxine be adequate because early development of the fetal brain depends on maternal thyroxine. In many hypothyroid patients, an increase in the thyroxine dose (about 25–30%) is required to normalize the serum TSH level during pregnancy. It is reasonable to counsel women to take an extra 25 mcg thyroxine tablet as soon as they are pregnant and to separate thyroxine from prenatal vitamins by at least 4 hours. Because of the elevated maternal TBG levels and, therefore, elevated total T4 levels, adequate maternal thyroxine dosages warrant maintenance of TSH between.0.1 and 3.0 mIU/L (eg, first trimester, 0.1–2.5 mIU/L; second trimester, 0.2–3.0 mIU/L; third trimester, 0.3–3.0 mIU/L) and the total T4 at or above the upper range of normal. D. Subclinical Hypothyroidism Subclinical hypothyroidism, defined as an elevated TSH level and normal thyroid hormone levels, occurs in 4–10% of the general population and increases to 20% in women older than age 50. The consensus of expert thyroid organizations concluded that thyroid hormone therapy should be considered for patients with TSH levels greater than 10 mIU/L while close TSH monitoring is appropriate for those with lower TSH elevations. E. Drug-Induced Hypothyroidism Drug-induced hypothyroidism (Table 38–3) can be satisfactorily managed with levothyroxine therapy if the offending agent cannot be stopped. In the case of amiodarone-induced hypothyroidism, levothyroxine therapy may be necessary even after discontinuance because of amiodarone’s very long half-life.

HYPERTHYROIDISM Hyperthyroidism (thyrotoxicosis) is the clinical syndrome that results when tissues are exposed to high levels of thyroid hormone (Table 38–4).


GRAVES’ DISEASE The most common form of hyperthyroidism is Graves’ disease, or diffuse toxic goiter. The presenting signs and symptoms of Graves’ disease are set forth in Table 38–4.

Pathophysiology Graves’ disease is considered to be an autoimmune disorder in which a defect in suppressor T lymphocytes stimulates B lymphocytes to synthesize antibodies to thyroidal antigens. The antibody described previously (TSH-R Ab [stim]) is directed against the TSH receptor site in the thyroid cell membrane and has the capacity to stimulate growth and biosynthetic activity of the thyroid cell. A genetic predisposition to Graves’ disease is shown by a high frequency of HLA-B8 and HLA-DR3 in Caucasians, HLA-Bw46 and HLA-B5 in Chinese, and HLA-B17 in African Americans. Spontaneous remission occurs but some patients require years of antithyroid therapy.

Laboratory Diagnosis In most patients with hyperthyroidism, T3 , T4 , FT4 , and FT3 are elevated and TSH is suppressed (Table 38–2). Radioiodine uptake is usually markedly elevated as well. Antithyroglobulin, thyroid peroxidase, and TSH-R Ab [stim] antibodies are usually present.

Management of Graves’ Disease The three primary methods for controlling hyperthyroidism are antithyroid drug therapy, surgical thyroidectomy, and destruction of the gland with radioactive iodine. A. Antithyroid Drug Therapy Drug therapy is most useful in young patients with small glands and mild disease. Methimazole (preferred) or propylthiouracil is administered until the disease undergoes spontaneous remission. This is the only therapy that leaves an intact thyroid gland, but it does require a long period of treatment and observation (12–18 months), and there is a 50–70% incidence of relapse. Methimazole is preferable to propylthiouracil (except in pregnancy and thyroid storm) because it has a lower risk of serious liver injury and can be administered once daily, which may improve adherence. Antithyroid drug therapy is usually begun with divided doses, shifting to maintenance therapy with single daily doses when the patient becomes clinically euthyroid. However, mild to moderately severe thyrotoxicosis can often be controlled with methimazole given in a single morning dose of 20–40 mg initially for 4–8 weeks to normalize hormone levels. Maintenance therapy requires 5–15 mg once daily. Alternatively, therapy is started with propylthiouracil, 100– 150 mg every 6 or 8 hours until the patient is euthyroid, followed by gradual reduction of the dose to the maintenance level of 50–150 mg once daily. In addition to inhibiting iodine organification, propylthiouracil also inhibits the conversion of T 4 to T3 , so it brings the level of activated thyroid hormone down more quickly than does methimazole. The best clinical guide to remission is reduction in the size of the goiter. Laboratory tests most useful in monitoring the course of therapy are serum FT3 , FT4 , and TSH levels. Reactions to antithyroid drugs have been described above. A minor rash can often be controlled by antihistamine therapy. Because the more severe reaction of agranulocytosis is often heralded by sore throat or high fever, patients receiving antithyroid drugs must be instructed to discontinue the drug and seek immediate medical attention if these symptoms develop. White cell and differential counts and a throat culture are indicated in such cases, followed by appropriate antibiotic therapy. Treatment should also be stopped if significant elevations in transaminases (two to three times the upper limit of normal) occur. B. Thyroidectomy A near-total thyroidectomy is the treatment of choice for patients with very large glands or multinodular goiters. Patients are treated with antithyroid drugs until euthyroid (about 6 weeks). In addition, for 10–14 days prior to surgery, they receive saturated solution of potassium iodide, 5 drops twice daily, to diminish vascularity of the gland and simplify surgery. About 80–90% of patients will require thyroid supplementation following near-total thyroidectomy. C. Radioactive Iodine Radioiodine therapy (RAI) utilizing 131 I is the preferred treatment for most patients over 21 years of age. In patients without heart disease, the therapeutic dose may be given immediately in a range of 80–120 μCi/g of estimated thyroid weight corrected for uptake. In patients with underlying heart disease or severe thyrotoxicosis and in elderly patients, it is desirable to treat with antithyroid drugs (preferably methimazole) until the patient is euthyroid. The medication is stopped for 3–5 days before RAI is administered so as not to interfere with RAI retention but can be restarted 3–7 days later, and then gradually tapered over 4–6 weeks as thyroid function normalizes. Iodides should be avoided to ensure maximal 131 I uptake. Six to 12 weeks following the administration of RAI, the gland will shrink in size and the patient will usually become euthyroid or hypothyroid. A second dose may be required if there is minimal response 3


months post-RAI. Hypothyroidism occurs in about 80% of patients following RAI. Serum FT4 and TSH levels should be monitored regularly. When hypothyroidism develops, prompt replacement with oral levothyroxine, 50–150 mcg daily, should be instituted. D. Adjuncts to Antithyroid Therapy During the acute phase of thyrotoxicosis, β-adrenoceptor–blocking agents without intrinsic sympathomimetic activity are appropriate in symptomatic patients aged 60 years or more, in those with heart rates greater than 90 beats/min, and in those with cardiovascular disease. Propranolol, 20–40 mg orally every 6 hours, or metoprolol, 25–50 mg orally every 6–8 hours, will control tachycardia, hypertension, and atrial fibrillation. Beta-adrenoceptor–blocking agents are gradually withdrawn as serum thyroxine levels return to normal. Diltiazem, 90–120 mg three or four times daily, can be used to control tachycardia in patients in whom β blockers are contraindicated, eg, those with asthma. Dihydropyridine calcium channel blockers may not be as effective as diltiazem or verapamil. Adequate nutrition and vitamin supplements are essential. Barbiturates accelerate T4 breakdown (by hepatic enzyme induction) and may be helpful both as sedatives and to lower T4 levels. Bile acid sequestrants (eg, cholestyramine) can also rapidly lower T4 levels by increasing the fecal excretion of T4 .

TOXIC UNINODULAR GOITER & TOXIC MULTINODULAR GOITER These forms of hyperthyroidism occur often in older women with nodular goiters. Free thyroxine is moderately elevated or occasionally normal, but FT3 or T3 is strikingly elevated. Single toxic adenomas can be managed with either surgical excision of the adenoma or with radioiodine therapy. Toxic multinodular goiter is usually associated with a large goiter and is best treated by preparation with methimazole (preferable) or propylthiouracil followed by subtotal thyroidectomy.

SUBACUTE THYROIDITIS During the acute phase of a viral infection of the thyroid gland, there is destruction of thyroid parenchyma with transient release of stored thyroid hormones. A similar state may occur in patients with Hashimoto’s thyroiditis. These episodes of transient thyrotoxicosis have been termed spontaneously resolving hyperthyroidism. Supportive therapy is usually all that is necessary, such as β-adrenoceptor– blocking agents without intrinsic sympathomimetic activity (eg, propranolol) for tachycardia and aspirin or nonsteroidal anti-inflammatory drugs to control local pain and fever. Corticosteroids may be necessary in severe cases to control the inflammation.

SPECIAL PROBLEMS Thyroid Storm Thyroid storm, or thyrotoxic crisis, is sudden acute exacerbation of all of the symptoms of thyrotoxicosis, presenting as a life-threatening syndrome. Vigorous management is mandatory. Propranolol, 60–80 mg orally every 4 hours, or intravenous propranolol, 1–2 mg slowly every 5-10 minutes to a total of 10 mg, or esmolol, 50–100 mg/kg/min, is helpful to control the severe cardiovascular manifestations. If β blockers are contraindicated by the presence of severe heart failure or asthma, hypertension and tachycardia may be controlled with diltiazem, 90–120 mg orally three or four times daily or 5–10 mg/h by intravenous infusion (asthmatic patients only). Release of thyroid hormones from the gland is retarded by the administration of saturated solution of potassium iodide, 5 drops orally every 6 hours starting 1 hour after giving thioamides. Hormone synthesis is blocked by the administration of propylthiouracil, 500–1000 mg as a loading dose, followed by 250 mg orally every 4 hours. If the patient is unable to take propylthiouracil by mouth, a rectal formulation* can be prepared and administered in a dosage of 400 mg every 6 hours as a retention enema. Methimazole may also be prepared for rectal administration in a dose of 60–80 mg daily. Hydrocortisone, 50 mg intravenously every 6 hours, will protect the patient against shock and will block the conversion of T4 to T3 , rapidly reducing the level of thyroactive material in the blood. Supportive therapy is essential to control fever, heart failure, and any underlying disease process that may have precipitated the acute storm. In rare situations, where the above methods are not adequate to control the problem, oral bile acid sequestrants (eg, cholestyramine), plasmapheresis, or peritoneal dialysis has been used to lower the levels of circulating thyroxine.

Ophthalmopathy Although severe ophthalmopathy is rare, it is difficult to treat. Exacerbations of severe eye disease may occur following RAI, especially in those who smoke. Management requires effective treatment of the thyroid disease, usually by total surgical excision or 131 I ablation of the gland plus oral prednisone therapy (see below). In addition, local therapy may be necessary, eg, elevation of the head to diminish periorbital edema and artificial tears to relieve corneal drying due to exophthalmos. Smoking cessation should be advised to prevent


progression of the ophthalmopathy. For the severe, acute inflammatory reaction, prednisone, 60–100 mg orally daily for about a week and then 60–100 mg every other day, tapering the dose over 6–12 weeks, may be effective. If steroid therapy fails or is contraindicated, irradiation of the posterior orbit, using well-collimated high-energy X-ray therapy, will frequently result in marked improvement of the acute process. Threatened loss of vision is an indication for surgical decompression of the orbit. Eyelid or eye muscle surgery may be necessary to correct residual problems after the acute process has subsided.

Dermopathy Dermopathy or pretibial myxedema will often respond to topical corticosteroids applied to the involved area and covered with an occlusive dressing.

Thyrotoxicosis during Pregnancy Ideally, women in the childbearing period with severe disease should have definitive therapy with 131 I or subtotal thyroidectomy prior to pregnancy in order to avoid an acute exacerbation of the disease during pregnancy or following delivery. If thyrotoxicosis does develop during pregnancy, RAI is contraindicated because it crosses the placenta and may injure the fetal thyroid. Propylthiouracil (fewer teratogenic risks than methimazole) can be given in the first trimester, and then methimazole can be given for the remainder of the pregnancy in order to avoid potential liver damage. The dosage of propylthiouracil must be kept to the minimum necessary for control of the disease (ie, < 300 mg/d), because it may affect the function of the fetal thyroid gland. Alternatively, a subtotal thyroidectomy can be safely performed during the mid trimester. It is essential to give the patient a thyroid supplement during the balance of the pregnancy.

Neonatal Graves’ Disease Graves’ disease may occur in the newborn infant, either due to passage of maternal TSH-R Ab [stim] through the placenta, stimulating the thyroid gland of the neonate, or to genetic transmission of the trait to the fetus. Laboratory studies reveal an elevated free T4 , a markedly elevated T3 , and a low TSH—in contrast to the normal infant, in whom TSH is elevated at birth. TSH-R Ab [stim] is usually found in the serum of both the child and the mother. If caused by maternal TSH-R Ab [stim], the disease is usually self-limited and subsides over a period of 4–12 weeks, coinciding with the fall in the infant’s TSH-R Ab [stim] level. However, treatment is necessary because of the severe metabolic stress the infant experiences. Therapy includes propylthiouracil at a dosage of 5–10 mg/kg/d in divided doses at 8-hour intervals; Lugol’s solution (8 mg of iodide per drop), 1 drop every 8 hours; and propranolol, 2 mg/kg/d in divided doses. Careful supportive therapy is essential. If the infant is very ill, oral prednisone, 2 mg/kg/d in divided doses, will help block conversion of T4 to T3 . These medications are gradually reduced as the clinical picture improves and can be discontinued by 6–12 weeks.

SUBCLINICAL HYPERTHYROIDISM Subclinical hyperthyroidism is defined as a suppressed TSH level (below the normal range) in conjunction with normal thyroid hormone levels. Cardiac toxicity (eg, atrial fibrillation), especially in older persons and those with underlying cardiac disease, is of greatest concern. The consensus of thyroid experts concluded that hyperthyroidism treatment is appropriate in those with TSH less than 0.1 mIU/L, while close monitoring of the TSH level is appropriate for those with less TSH suppression.

Amiodarone-Induced Thyrotoxicosis In addition to those patients who develop hypothyroidism caused by amiodarone, approximately 3% of patients receiving this drug will develop hyperthyroidism instead. Two types of amiodarone-induced thyrotoxicosis have been reported: iodine-induced (type I), which often occurs in persons with underlying thyroid disease (eg, multinodular goiter, Graves’ disease); and an inflammatory thyroiditis (type II) that occurs in patients without thyroid disease due to leakage of thyroid hormone into the circulation. Treatment of type I requires therapy with thioamides, while type II responds best to glucocorticoids. Since it is not always possible to differentiate between the two types, thioamides and glucocorticoids are often administered together. If possible, amiodarone should be discontinued; however, rapid improvement does not occur due to its long half-life.

NONTOXIC GOITER Nontoxic goiter is a syndrome of thyroid enlargement without excessive thyroid hormone production. Enlargement of the thyroid gland is often due to TSH stimulation from inadequate thyroid hormone synthesis. The most common cause of nontoxic goiter worldwide is iodide deficiency, but in the USA, it is Hashimoto’s thyroiditis. Other causes include germ-line or acquired mutations in genes involved in hormone synthesis, dietary goitrogens, and neoplasms (see below).


Goiter due to iodide deficiency is best managed by prophylactic administration of iodide. The optimal daily iodide intake is 150–200 mcg. Iodized salt and iodate used as preservatives in flour and bread are excellent sources of iodine in the diet. In areas where it is difficult to introduce iodized salt or iodate preservatives, a solution of iodized poppy-seed oil has been administered intramuscularly to provide a long-term source of inorganic iodine. Goiter due to ingestion of goitrogens in the diet is managed by elimination of the goitrogen or by adding sufficient thyroxine to shut off TSH stimulation. Similarly, in Hashimoto’s thyroiditis and dyshormonogenesis, adequate thyroxine therapy—150–200 mcg/d orally—will suppress pituitary TSH and result in slow regression of the goiter as well as correction of hypothyroidism.

THYROID NEOPLASMS Neoplasms of the thyroid gland may be benign (adenomas) or malignant. The primary diagnostic test is a fine needle aspiration biopsy and cytologic examination. Benign lesions may be monitored for growth or symptoms of local obstruction, which would mandate surgical excision. Levothyroxine therapy is not recommended for the suppression of benign nodules, especially in iodine sufficient areas. Management of thyroid carcinoma requires a total thyroidectomy, postoperative radioiodine therapy in selected instances, and lifetime replacement with levothyroxine. The evaluation for recurrence of some thyroid malignancies often involves withdrawal of thyroxine replacement for 4–6 weeks—accompanied by the development of hypothyroidism. Tumor recurrence is likely if there is a rise in serum thyroglobulin (ie, a tumor marker) or a positive 131 I scan when TSH is elevated. Alternatively, administration of recombinant human TSH (Thyrogen) can produce comparable TSH elevations without discontinuing thyroxine and avoiding hypothyroidism. Recombinant human TSH is administered intramuscularly once daily for 2 days. A rise in serum thyroglobulin or a positive 131 I scan will indicate a recurrence of the thyroid cancer.

SUMMARY Drugs Used in the Management of Thyroid Disease



PREPARATIONS AVAILABLE

REFERENCES General American T hyroid Association T ask Force On Radiation Safety et al: Radiation safety in the treatment of patients with thyroid diseases by radioiodine 131I: Practice recommendations of the American T hyroid Association. T hyroid 2011;21:335 (http://thyroidguidelines.net/sites/thyroidguidelines.net/files/file/thy.2010.0403.pdf). Bahn RS et al: Hyperthyroidism and other causes of thyrotoxicosis: Management guidelines of the American T hyroid Association and American Association of Clinical Endocrinologists. T hyroid 2011;21:593 (http://thyroidguidelines.net/sites/thyroidguidelines.net/files/file/T HY_2010_0417.pdf).


Biondi B: Natural history, diagnosis, and management of subclinical thyroid dysfunction. Best Pract Res Clin Endocrinol Metab 2012;26:431. Bochukova E et al: A mutation in the thyroid hormone receptor alpha gene. N Engl J Med 2012;366:243. Cooper DS: T he clinical significance of subclinical thyroid dysfunction. Endocr Rev 2008;29:76. Cooper DS et al: Revised American T hyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. T hyroid 2009;19:1167. Cooper DS et al: T he thyroid gland. In: Gardner DG, Shoback D (editors): Greenspan’s Basic & Clinical Endocrinology, 9th ed. McGraw-Hill, 2011. Galli E, Pingitore A, Iervasi G: T he role of thyroid hormone in the pathophysiology of heart failure: Clinical evidence. Heart Fail Rev 2010;15:155. Garber JR et al: American Association of Clinical Endocrinologists and American T hyroid Association T askforce on Hypothyroidism in Adults. T hyroid 2012;22:1200 (http://www.thyroidguidelines.net/sites/thyroidguidelines.net/files/file/thy.2012.0205.pdf). Guidelines of the American T hyroid Association (http://www.thyroid.org). Laurberg P et al: Iodine intake as a determinant of thyroid disorders in populations. Best Pract Res Clin Endocrinol Metab 2010;24:13. Negro R, Mestman J: T hyroid disease in pregnancy. Best Pract Res Clin Endocrinol Metab 2011;25:927. Oetting A, Yen PM: New insights into thyroid hormone action. Best Pract Res Clin Endocrinol Metab 2007;21:193. Porcu E et al: A meta-analysis of thyroid-related traits reveals novel loci and gender-specific differences in the regulation of thyroid function. PLoS Genet 2013;9:e1003266. doi:10.1371/journal.pgen.1003266. Epub 2013 Feb 7. Stagnaro-Green A et al: T he American T hyroid Association T askforce on T hyroid Disease During Pregnancy and Postpartum. T hyroid 2011;21:1081 (http://thyroidguidelines.net/sites/thyroidguidelines.net/files/file/thy.2011.0087.pdf). Ross DS: T reatment of hypothyroidism. Up to Date 2013; http://www.uptodate.com/contents/search? sp=0&source=USER_PREF&search=thyroid&searchT ype=PLAIN_T EXT . US Department of Health and Human Services: Potassium iodide as a thyroid blocking agent in radiation emergencies. December 2001. Available at: http://www.fda.gov/cder/guidance/index.htm. Williams GR: Neurodevelopment and neurophysiological actions of thyroid hormone. J Neuroendocrinol 2008;20:784.

Hypothyroidism Dong BJ et al: Bioequivalence of generic and brand-name levothyroxine products in the treatment of hypothyroidism. JAMA 1997;277:1205. Hoang T D et al: Desiccated thyroid extract compared with levothyroxine in the treatment of hypothyroidism: A randomized, double-blind, crossover study. J Clin Endocrinol Metab 2013;98:1982. Joffe RT et al: T reatment of clinical hypothyroidism with thyroxine and triiodothyronine: A literature review and meta-analysis. Psychosomatics 2007;48:379. Jonklaas J et al: T riiodothyronine levels in athyreotic individuals during levothyroxine therapy. JAMA 2008;299:769. Panicker V et al: Common variation in the DIO2 gene predicts baseline psychological well-being and response to combination thyroxine plus triiodothyronine therapy in hypothyroid patients. J Clin Endocrinol Metab 2009;94:1623. Vita R et al: A novel formulation of L-thyroxine (L-T 4) reduces the problem of L-T 4 malabsorption by coffee observed with traditional tablet formulations. Endocrine 2013;43:154. Wartofsky L: Combination L-T 3 and L-T 4 therapy for hypothyroidism. Curr Opin Endocrinol Diabetes Obes. 2013;20:460.

Hyperthyroidism Abraham P et al: Antithyroid drug regimen for treating Graves’ hyperthyroidism. Cochrane Database Syst Rev 2010;(1):CD003420 (http://onlinelibrary.wiley.com/o/cochrane/clsysrev/articles/CD003420/pdf_fs.html). Bahn RS: Graves’ ophthalmopathy. N Engl J Med 2010;362:726. Brent GA: Graves’ disease. N Engl J Med 2008;358:2594. Cooper DS, Rivkees SA: Putting propylthiouracil in perspective. J Clin Endocrinol Metab 2009;94:1881. Hegedüs L et al: T reating the thyroid in the presence of Graves’ ophthalmopathy. Best Pract Res Clin Endocrinol Metab 2012; 26:313–324 (June 2012 entire issue devoted to Grave’ ophthalmopathy diagnosis and management). Silva JE, Bianco SD: T hyroid-adrenergic interactions: Physiological and clinical implications. T hyroid 2008;18:157. Sundaraesh V et al. Comparative Effectiveness of T herapies for Graves’ Hyperthyroidism: A Systematic Review and Network Meta-Analysis. J Clin Endo Metab 2013 98: 367.

Nodules & Cancer (see Guidelines) Gharib H et al: Clinical review: Nonsurgical, image-guided, minimally invasive therapy for thyroid nodules. J Clin Endocrinol Metab 2013;98:3949.

The Effects of Drugs on Thyroid Function Barbesino G: Drugs affecting thyroid function. T hyroid 2010;20:763. Burgi H: Iodine excess. Best Pract Res Clin Endocrinol Metab 2010;24:107. Eskes SA, Wiersinga WM: Amiodarone and the thyroid. Best Pract Res Clin Endocrinol Metab 2009;23:735. Haugen BR: Drugs that suppress T SH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab 2009;23:793. Lazarus JH: Lithium and thyroid. Best Pract Res Clin Endocrinol Metab 2009;23:723. Makita N, Liri T : T yrosine kinase inhibitor–induced thyroid disorders: A review and hypothesis. T hyroid 2013;23:151.


Mammen JS et al: Patterns of interferon-alpha–induced thyroid dysfunction vary with ethnicity, sex, smoking status, and pretreatment thyrotropin in an international cohort of patients treated for hepatitis C. T hyroid 2013;23:1151. T omer Y, Menconi F: Interferon induced thyroiditis. Best Pract Res Clin Endocrinol Metab 2009;23:703.

CASE STUDY ANSWER This patient presents with the typical signs and symptoms of hypothyroidism following radioactive iodine therapy. Radioactive iodine therapy and thyroidectomy are reasonable and effective strategies for definitive treatment of her hyperthyroidism, especially before becoming pregnant to avoid an acute exacerbation of the disease during pregnancy or following delivery. This patient’s hypothyroid symptoms are easily corrected by the daily administration of levothyroxine, taken orally 60 minutes before meals on an empty stomach. Thyroid function tests should be checked after 6–8 weeks, before thyroxine administration to avoid transient hormone alterations, and the dosage adjusted to achieve a normal TSH level and resolution of hypothyroid symptoms.


*

To prepare a water suspension propylthiouracil enema, grind eight 50 mg tablets and suspend the powder in 90 mL of sterile water.


Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.