CHAPTER 43 is produced by myocytes of the atria; BNP is produced by myocytes of the ventricles (and to a lesser extent by cells in the brain, where BNP was discovered); and CNP is produced by cells of the vascular endothelium. When blood volume is excessive, all three peptides are released. (Release of ANP and BNP is triggered by stretching of the atria and ventricles, which occurs because of increased preload.) ANP and BNP have similar actions. Both peptides reduce blood volume and increase venous capacitance, and thereby reduce cardiac preload. Three processes are involved. First, ANP and BNP shift fluid from the vascular system to the Review of Hemodynamics extravascular compartment; the underlying mechanism is increased vascular permeability. Second, these peptides act on the kidney to cause diuresis (loss of water) and natriuresis (loss of sodium). Third, they promote dilation of arterioles and veins, in part by suppressing sympathetic outflow from the central nervous system. In addition to these actions, ANP and BNP help protect the heart during the early phase of heart failure by suppressing both the RAAS and sympathetic outflow, and by inhibiting proliferation of myocytes. Although CNP shares some actions of ANP and BNP, its primary action is to promote vasodilation. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Arterioles serve as control valves to regulate local blood flow. Veins are a reservoir for blood. Arteries are not very distensible. As a result, large increases in arterial pressure (AP) cause only small increases in arterial diameter. Veins are highly distensible. As a result, small increases in venous pressure cause large increases in venous diameter. The adult circulatory system contains 5 L of blood, 64% of which is in systemic veins. Vasodilation reduces resistance to blood flow, whereas vasoconstriction increases resistance to flow. In addition to the small pressure head in venules, three mechanisms help ensure venous return to the heart: (1) negative pressure in the right atrium sucks blood toward the heart; (2) constriction of veins increases venous pressure and thereby drives blood toward the heart; and (3) contraction of skeletal muscles, in conjunction with one-way venous valves, pumps blood toward the heart. Heart rate is increased by sympathetic nerve impulses and decreased by parasympathetic impulses. Stroke volume is determined by myocardial contractility, cardiac preload, and cardiac afterload. Preload is defined as the amount of tension (stretch) applied to a muscle before contraction. In the heart, preload is determined by the force of venous return. Afterload is defined as the load against which a muscle exerts its force. For the heart, afterload is the AP that the left ventricle must overcome to eject blood. Cardiac afterload is determined primarily by peripheral resistance, which in turn is determined by the degree of constriction in arterioles. Starling’s law states that the force of ventricular contraction is proportional to myocardial fiber length. Because of this relationship, when more blood enters the heart, more is pumped out. As a result, the healthy heart is able to precisely match output with venous return. The most important determinant of venous return is systemic filling pressure, which can be raised by constricting veins and increasing blood volume. ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Because cardiac muscle operates under Starling’s law, the right and left ventricles always pump exactly the same amount of blood (assuming the heart is healthy). Hence, balance between the pulmonary and systemic circulations is maintained. Arterial pressure is regulated by the ANS, the RAAS, the kidneys, and natriuretic peptides. The ANS regulates AP (1) through tonic control of heart rate and peripheral resistance and (2) through the baroreceptor reflex. The baroreceptor reflex is useful only for short-term control of AP. When pressure remains elevated or lowered, the system resets to the new pressure within 1 to 2 days, and hence ceases to respond. Drugs that lower AP trigger the baroreceptor reflex and thereby cause reflex tachycardia. Hence, the baroreceptor reflex can temporarily negate efforts to lower AP with drugs. The RAAS supports AP by causing (1) constriction of arterioles and veins and (2) retention of water by the kidneys. Vasoconstriction is mediated by angiotensin II; water retention is mediated in part by aldosterone. The kidneys provide long-term control of blood pressure by regulating blood volume. Postural (orthostatic) hypotension is caused by decreased venous return secondary to pooling of blood in veins, which can occur when we assume an erect posture. Drugs that dilate veins intensify and prolong postural hypotension. As with other drugs that reduce AP, venodilators can trigger the baroreceptor reflex and can thereby cause reflex tachycardia. Natriuretic peptides defend the cardiovascular system from volume overload—primarily by reducing blood volume and promoting vasodilation. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. 481 CHAPTER 44 unknown. Eplerenone undergoes metabolism by CYP3A4, followed by excretion in the urine (67%) and feces (32%). The elimination half-life is 4 to 6 hours. Adverse Effects Eplerenone is generally well tolerated. The incidence of adverse effects is nearly identical to that of placebo. A few adverse effects—diarrhea, abdominal pain, cough, fatigue, gynecomastia, flu-like syndrome—occur slightly (1% to 2%) more often with eplerenone than with placebo. Hyperkalemia. The greatest concern is hyperkalemia, which can occur secondary to potassium retention. Because of this risk, combined use with potassium supplements, salt substitutes, or potassium-sparing diuretics (e.g., spironolactone, triamterene) is contraindicated. Combined use with ACE inhibitors or ARBs is permissible, but should be done with caution. Eplerenone is contraindicated for patients with high serum potassium (above 5.5 mEq/L) and for patients with impaired renal function or type 2 diabetes with microalbuminuria, both of which can promote hyperkalemia. Monitoring potassium levels is recommended for patients at risk (e.g., those taking ACE inhibitors or ARBs). Drug Interactions Inhibitors of CYP3A4 can increase levels of eplerenone, thereby posing a risk of toxicity. Weak inhibitors (e.g., erythromycin, saquinavir, verapamil, fluconazole) can double eplerenone levels. Strong inhibitors (e.g., ketoconazole, itraconazole) can increase levels fivefold. If eplerenone is combined with a weak inhibitor, eplerenone dosage should be reduced. Eplerenone should not be combined with a strong inhibitor. Drugs Acting on the Renin-Angiotensin-Aldosterone System Drugs that raise potassium levels can increase the risk of hyperkalemia. Eplerenone should not be combined with potassium supplements, salt substitutes, or potassium-sparing diuretics. Combining eplerenone with ACE inhibitors or ARBs should be done with caution. Drugs similar to eplerenone (e.g., ACE inhibitors and diuretics) are known to increase levels of lithium. Although the combination of eplerenone and lithium has not been studied, caution is nonetheless advised. Lithium levels should be measured frequently. Preparations, Dosage, and Administration Eplerenone [Inspra] is available in 25- and 50-mg tablets. The usual starting dosage is 50 mg once a day, taken with or without food. After 4 weeks, dosage can be increased to 50 mg twice daily (if the hypotensive response has been inadequate). Raising the dosage above 100 mg/day is not recommended because doing so is unlikely to increase the therapeutic response, but will increase the risk of hyperkalemia. In patients taking weak inhibitors of CYP3A4, the initial dosage should be reduced by 50% (to 25 mg once a day). Spironolactone Spironolactone [Aldactone], a much older drug than eplerenone, blocks receptors for aldosterone, but also binds with receptors for other steroid hormones (e.g., glucocorticoids, progesterone, androgens). Blockade of aldosterone receptors underlies beneficial effects in hypertension and heart failure, as well as the drug’s major adverse effect: hyperkalemia. Binding with receptors for other steroid hormones underlies additional adverse effects: gynecomastia, menstrual irregularities, impotence, hirsutism, and deepening of the voice. The basic pharmacology of spironolactone and its use in heart failure are discussed in Chapters 41 and 48, respectively. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ The RAAS helps regulate blood pressure, blood volume, and fluid and electrolyte balance. The system can promote cardiovascular pathology. The RAAS acts through production of angiotensin II and aldosterone. Angiotensin II has much greater biologic activity than angiotensin I or angiotensin III. Angiotensin II is formed by the actions of two enzymes: renin and ACE. Angiotensin II causes vasoconstriction (primarily in arterioles) and release of aldosterone. In addition, angiotensin II can promote pathologic changes in the heart and blood vessels. Aldosterone acts on the kidneys to promote retention of sodium and water. In addition, aldosterone can also mediate pathologic changes in cardiovascular function. The RAAS raises blood pressure by causing vasoconstriction and by increasing blood volume (secondary to aldosteronemediated retention of sodium and water). In addition to the traditional RAAS, in which angiotensin II is produced in the blood and then carried to target tissues, angiotensin II can be produced locally by individual tissues. Beneficial effects of ACE inhibitors result largely from inhibition of ACE and partly from inhibition of kinase II (the name for ACE when the substrate is bradykinin). ■ ■ ■ ■ ■ ■ ■ By inhibiting ACE, ACE inhibitors decrease production of angiotensin II. The result is vasodilation, decreased blood volume, and prevention or reversal of pathologic changes in the heart and blood vessels mediated by angiotensin II and aldosterone. ACE inhibitors (and ARBs) are used to treat patients with hypertension, heart failure, MI, and established diabetic nephropathy. In addition, they are used to prevent MI, stroke, and death from cardiovascular causes in patients at high risk for a cardiovascular event. Of note, ACE inhibitors (and ARBs) are not effective for primary prevention of diabetic nephropathy. Preliminary data indicate that ACE inhibitors (and ARBs) can reduce the risk of developing diabetic retinopathy, although they can’t slow the progression of established retinopathy. ACE inhibitors can produce significant first-dose hypotension by causing a sharp drop in circulating angiotensin II. Cough, secondary to accumulation of bradykinin, is the most common reason for discontinuing ACE inhibitors. By suppressing aldosterone release, ACE inhibitors can cause hyperkalemia. Exercise caution in patients taking potassium supplements, salt substitutes, or potassiumsparing diuretics. ACE inhibitors can cause major fetal malformations and should be avoided during pregnancy. Until recently, Continued 493 UNIT VII ■ ■ ■ ■ ■ Drugs That Affect the Heart, Blood Vessels, and Blood we thought that risk was limited to exposure during the second and third trimesters. However, new data indicate that exposure during the first trimester may be dangerous as well. ACE inhibitors can cause a precipitous drop in blood pressure in patients with bilateral renal artery stenosis (or stenosis in the artery to a single remaining kidney). ARBs block the actions of angiotensin II in blood vessels, the adrenals, and all other tissues. ARBs are similar to ACE inhibitors in that they cause vasodilation, suppress aldosterone release, promote excretion of sodium and water, reduce blood pressure, and cause birth defects and angioedema. ARBs differ from ACE inhibitors in that they have a much lower incidence of hyperkalemia or cough. Aliskiren, a DRI, binds tightly with renin and thereby inhibits cleavage of angiotensinogen into angiotensin I. As a result, the drug suppresses the entire RAAS. ■ ■ ■ ■ Like the ACE inhibitors and ARBs, aliskiren causes vasodilation, suppresses aldosterone release, promotes excretion of sodium and water, reduces blood pressure, and causes birth defects and angioedema. Despite their similarities, aliskiren, ARBs, and ACE inhibitors are not clinically interchangeable. Aldosterone antagonists (spironolactone, eplerenone) block receptors for aldosterone. By blocking aldosterone receptors, aldosterone antagonists can (1) promote renal excretion of sodium and water (and can thereby reduce blood volume and blood pressure) and (2) prevent or reverse pathologic effects of aldosterone on cardiovascular structure and function. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa Identifying High-Risk Patients ACE inhibitors are contraindicated during the second and third trimesters of pregnancy and for patients with (1) bilateral renal artery stenosis (or stenosis in the artery to a single remaining kidney) or (2) a history of hypersensitivity reactions (especially angioedema) to ACE inhibitors. Exercise caution in patients with salt or volume depletion, renal impairment, or collagen vascular disease, and in those taking potassium supplements, salt substitutes, potassiumsparing diuretics, ARBs, aliskiren, or lithium. ANGIOTENSIN-CONVERTING ENZYME INHIBITORS Benazepril Captopril Enalapril Enalaprilat Fosinopril Lisinopril Moexipril Perindopril Quinapril Ramipril Trandolapril Unless indicated otherwise, the implications summarized in the sections that follow pertain to all of the ACE inhibitors. Preadministration Assessment Therapeutic Goals ACE inhibitors are used to: Implementation: Administration Routes Oral. All ACE inhibitors (except enalaprilat). Intravenous. Enalaprilat. Dosage and Administration Dosage is low initially and then gradually increased. Instruct patients to administer captopril and moexipril at least 1 hour before meals. All other oral ACE inhibitors can • Reduce blood pressure in patients with hypertension (all ACE inhibitors). • Improve hemodynamics in patients with heart failure (captopril, enalapril, fosinopril, lisinopril, quinapril). • Slow progression of established diabetic nephropathy (captopril). • Reduce mortality following acute MI (lisinopril). • Treat heart failure after MI (ramipril, trandolapril). • Reduce the risk of MI, stroke, or death from cardiovascular causes in patients at high risk (ramipril). • Reduce cardiovascular mortality or nonfatal MI in patients with stable coronary artery disease (perindopril). Baseline Data Determine blood pressure, and obtain a white blood cell count and differential. be administered with food. Ongoing Evaluation and Interventions Monitoring Summary Monitor blood pressure closely for 2 hours after the first dose and periodically thereafter. Obtain a white blood cell count and differential every 2 weeks for the first 3 months of therapy and periodically thereafter. Evaluating Therapeutic Effects Hypertension. Monitor for reduced blood pressure. The usual target pressure is systolic/diastolic of 140/90 mm Hg or 130/80 in patients with diabetes. Heart Failure. Monitor for a lessening of signs and symptoms (e.g., dyspnea, cyanosis, jugular vein distention, edema). 494 CHAPTER 44 Drugs Acting on the Renin-Angiotensin-Aldosterone System Summary of Major Nursing Implicationsa—cont’d Diabetic Nephropathy. Monitor for proteinuria and altered glomerular filtration rate. Minimizing Adverse Effects First-Dose Hypotension. Severe hypotension can occur with the first dose. Minimize hypotension by (1) withdrawing diuretics 2 to 3 days before initiating ACE inhibitors and (2) using low initial doses. Monitor blood pressure for 2 hours following the first dose. Instruct patients to lie down if hypotension develops. If necessary, infuse normal saline to restore pressure. Cough. Warn patients about the possibility of persistent dry, irritating, nonproductive cough. Instruct them to consult the prescriber if cough is bothersome. Stopping the ACE inhibitor may be indicated. Hyperkalemia. ACE inhibitors may increase potassium levels. Instruct patients to avoid potassium supplements and potassium-containing salt substitutes unless they are prescribed by the provider. Potassium-sparing diuretics must also be avoided. Fetal Injury. Warn women of childbearing age that taking ACE inhibitors during the second and third trimesters of pregnancy can cause major fetal injury (hypotension, hyperkalemia, skull hypoplasia, anuria, reversible and irreversible renal failure, death) and that taking these drugs earlier in pregnancy may pose a risk as well. If the patient becomes pregnant, withdraw ACE inhibitors as soon as possible. Closely monitor infants who have been exposed to ACE inhibitors during the second or third trimester for hypotension, oliguria, and hyperkalemia. Angioedema. This rare and potentially fatal reaction is characterized by giant wheals and edema of the tongue, glottis, and pharynx. Instruct patients to seek immediate medical attention if these symptoms develop. If angioedema is diagnosed, ACE inhibitors should be discontinued and never used again. Treat severe reactions with subcutaneous epinephrine. Renal Failure. Renal failure is a risk for patients with bilateral renal artery stenosis or stenosis in the artery to a single remaining kidney. ACE inhibitors must be used with extreme caution in these people. Neutropenia (Mainly With Captopril). Neutropenia poses a high risk of infection. Inform patients about early signs of infection (fever, sore throat, mouth sores), and instruct them to notify the prescriber if these occur. If neutropenia develops, withdraw the drug immediately; neutrophil counts should normalize in approximately 2 weeks. Neutropenia is most likely in patients with renal impairment and collagen vascular diseases (e.g., systemic lupus erythematosus, scleroderma); monitor these patients closely. Minimizing Adverse Interactions Diuretics. Diuretics may intensify first-dose hypotension. Withdraw diuretics 2 to 3 days before beginning an ACE inhibitor. Diuretics may be resumed later if needed. Antihypertensive Agents. The antihypertensive effects of ACE inhibitors are additive with those of other antihypertensive drugs (e.g., ARBs, diuretics, sympatholytics, vasodilators, calcium channel blockers). When an ACE inhibitor is added to an antihypertensive regimen, dosages of the other drugs may require reduction. Drugs That Elevate Potassium Levels. ACE inhibitors increase the risk of hyperkalemia associated with potassium supplements, potassium-sparing diuretics, and possibly aliskiren. Risk can be minimized by avoiding potassium supplements and potassium-sparing diuretics except when they are clearly indicated. Lithium. ACE inhibitors can increase serum levels of lithium, causing toxicity. Monitor lithium levels frequently. Nonsteroidal Anti-Inflammatory Drugs. NSAIDs (e.g., aspirin, ibuprofen) can interfere with the antihypertensive effects of ACE inhibitors. Advise patients to minimize NSAID use. ANGIOTENSIN II RECEPTOR BLOCKERS Azilsartan Candesartan Eprosartan Irbesartan Losartan Olmesartan Telmisartan Valsartan Unless indicated otherwise, the implications summarized here pertain to all of the ARBs. Preadministration Assessment Therapeutic Goals ARBs are used to: • Reduce blood pressure in patients with hypertension (all ARBs). • Treat heart failure (candesartan, valsartan). • Slow the progression of established diabetic nephropathy (irbesartan, losartan). • Prevent stroke in patients with hypertension and LV hypertrophy (losartan). • Protect against MI, stroke, and death from cardiovascular causes in high-risk patients, but only if they can’t tolerate ACE inhibitors (telmisartan). • Treat heart failure after MI (valsartan). Baseline Data Determine blood pressure. Identifying High-Risk Patients ARBs are contraindicated during the second and third trimesters of pregnancy and for patients with either (1) bilateral renal artery stenosis (or stenosis in the artery to a single remaining kidney) or (2) a history of hypersensitivity reactions (especially angioedema) to ARBs. Implementation: Administration Route Oral. Continued 495 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d Dosage and Administration Inform patients that ARBs may be taken with or without food. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Hypertension. Monitor for reduced blood pressure. The usual target pressure is systolic/diastolic of 140/90 mm Hg or 130/80 in patients with diabetes. Heart Failure. Monitor for a lessening of signs and symptoms (e.g., dyspnea, cyanosis, jugular vein distention, edema). Diabetic Nephropathy. Monitor for proteinuria and altered glomerular filtration rate. Minimizing Adverse Effects Angioedema. This rare and potentially fatal reaction is characterized by giant wheals and edema of the tongue, glottis, and pharynx. Instruct patients to seek immediate medical attention if these symptoms develop. If angioedema is diagnosed, ARBs should be discontinued and never used again. Treat severe reactions with subcutaneous epinephrine. Fetal Injury. Warn women of childbearing age that ARBs can cause fetal injury during the second and third trimesters of pregnancy, and may pose a risk earlier in pregnancy as well. If the patient becomes pregnant, withdraw ARBs as soon as possible. Closely monitor infants who have been exposed to ARBs during the second or third trimester for hypotension, oliguria, and hyperkalemia. Renal Failure. Renal failure is a risk for patients with bilateral renal artery stenosis or stenosis in the artery to a single remaining kidney. ARBs are contraindicated for these people. Minimizing Adverse Interactions Antihypertensive Agents. The antihypertensive effects of ARBs are additive with those of other antihypertensive drugs (e.g., diuretics, sympatholytics, vasodilators, ACE inhibitors, calcium channel blockers). When an ARB is added to an antihypertensive regimen, dosages of the other drugs may require reduction. ALISKIREN, A DIRECT RENIN INHIBITOR Preadministration Assessment Therapeutic Goal Reduction of blood pressure in patients with hypertension. Baseline Data Determine blood pressure. Exercise caution in patients taking potassium supplements, salt substitutes, potassium-sparing diuretics, or ACE inhibitors. Implementation: Administration Route Oral. Dosage and Administration Advise patients to take each daily dose at the same time with respect to meals (e.g., 1 hour before dinner). Dosage should be low (150 mg/day) initially, and increased to a maximum of 300 mg/day if needed. Ongoing Evaluation and Interventions Minimizing Adverse Effects Hyperkalemia. Aliskiren may increase potassium levels. Instruct patients to avoid potassium supplements and potassium-containing salt substitutes unless they are prescribed by the provider. Potassium-sparing diuretics must also be avoided. Exercise caution in patients taking an ACE inhibitor. Fetal Injury. Warn women of childbearing age that aliskiren taken during the second and third trimesters of pregnancy can cause fetal injury (hypotension, hyperkalemia, skull hypoplasia, anuria, reversible and irreversible renal failure, death). If the patient becomes pregnant, withdraw aliskiren as soon as possible. Closely monitor infants who have been exposed to aliskiren during the second or third trimester for hypotension, oliguria, and hyperkalemia. Angioedema. This rare and potentially fatal reaction is characterized by giant wheals and edema of the tongue, glottis, and pharynx. Instruct patients to seek immediate medical attention if these symptoms develop. If angioedema is diagnosed, aliskiren should be discontinued and never used again. Treat severe reactions with subcutaneous epinephrine. Minimizing Adverse Interactions Drugs That Elevate Potassium Levels. Aliskiren increases the risk of hyperkalemia associated with ACE inhibitors, potassium supplements, and potassium-sparing diuretics. Risk can be minimized by avoiding ACE inhibitors, potassium supplements, and potassium-sparing diuretics except when they are clearly indicated. Antihypertensive Agents. The antihypertensive effects of aliskiren are additive with those of other antihypertensive drugs (e.g., ACE inhibitors, ARBs, diuretics, sympatholytics, vasodilators, calcium channel blockers). When aliskiren is added to an antihypertensive regimen, dosages of the other drugs may require reduction. Identifying High-Risk Patients Aliskiren is contraindicated during the second and third trimesters of pregnancy. Patient education information is highlighted as blue text. a 496 CHAPTER 45 of neurologic injury following rupture of an intracranial aneurysm. Benefits derive from preventing cerebral arterial spasm that follows subarachnoid hemorrhage (SAH) and can result in ischemic neurologic injury. Dosing (60 mg every 4 hours) should begin within 96 hours of SAH and continue for 21 days. Nimotop is available in 30-mg liquid-filled capsules for oral administration and as a 60 mg/20 mL oral solution [Nymalize]. Nimodipine must never be given intravenously, owing to a risk of potentially fatal cardiovascular events. Nisoldipine Like nifedipine, nisoldipine [Sular] produces selective blockade of calcium channels in blood vessels; the drug has minimal direct effects on the heart. The only approved indication is hypertension. Nisoldipine is well absorbed following oral administration, but the first-pass effect limits bioavailability to 5%. Plasma levels peak 6 hours after administration. The most common side effects are dizziness, headache, and peripheral edema. Reflex tachycardia may also occur. As with other CCBs, eczematous rash may develop in older patients. Nisoldipine is available in ER tablets (8.5, 17, 20, 25.5, 30, 34, and 40 mg). The dosage for hypertension is 17 to 60 mg once a day. Calcium Channel Blockers Clevidipine Clevidipine [Cleviprex] is indicated only for intravenous therapy of severe hypertension, defined as systolic blood pressure above 180 mm Hg or diastolic pressure above 110 mm Hg. The drug has an ultrashort half-life (about 1 minute), owing to rapid inactivation by plasma esterases. Effects are not altered by impairment of liver or kidney function. Because of IV dosing and rapid inactivation, blood pressure falls quickly and then rises quickly when the infusion is slowed or stopped. As a result, responses can be easily titrated. Clevidipine is formulated in a lipid emulsion made from soybean oil and egg yolk phospholipids, and hence is contraindicated for patients allergic to soybeans or eggs. Clevidipine is supplied in single-dose vials (50, 100, or 250 mL) at a concentration of 0.5 mg/mL. For patients with severe hypertension, the infusion rate is 1 to 2 mg/hr initially, and can be doubled every 90 seconds up to a maximum of 32 mg/hr. In clinical trials, the average time to reach the target blood pressure was 10.9 minutes. The most common side effects are headache, nausea, and vomiting. Like other dihydropyridines, clevidipine can cause hypotension and reflex tachycardia. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ Calcium channels are gated pores in the cytoplasmic membrane that regulate calcium entry into cells. In blood vessels, calcium entry causes vasoconstriction, and hence calcium channel blockade causes vasodilation. In the heart, calcium entry increases heart rate, AV conduction, and myocardial contractility, so calcium channel blockade has the opposite effects. In the heart, calcium channels are coupled to beta1 receptors, activation of which enhances calcium entry. As a result, calcium channel blockade and beta blockade have identical effects on cardiac function. At therapeutic doses, nifedipine and the other dihydropyridines act primarily on VSM; in contrast, verapamil and diltiazem act on VSM and on the heart. All CCBs promote vasodilation, and hence are useful in hypertension and angina pectoris. Because they suppress AV conduction, verapamil and diltiazem are useful for treating cardiac dysrhythmias (in addition to hypertension and angina pectoris). Because of their cardiosuppressant effects, verapamil and diltiazem can cause bradycardia, partial or complete AV block, and exacerbation of heart failure. ■ ■ ■ ■ ■ ■ Beta blockers intensify cardiosuppression caused by verapamil and diltiazem. Nifedipine and other dihydropyridines can cause reflex tachycardia. Tachycardia is most intense with immediaterelease formulations, and much less intense with sustainedrelease formulations. Beta blockers can be used to suppress reflex tachycardia caused by nifedipine and other dihydropyridines. Because they cause vasodilation, all CCBs can cause dizziness, headache, and peripheral edema. In toxic doses, nifedipine and other dihydropyridines can cause cardiosuppression, just like verapamil and diltiazem. Immediate-release nifedipine has been associated with increased mortality in patients with myocardial infarction and unstable angina, although a causal relationship has not been established. The National Heart, Lung, and Blood Institute recommends that immediate-release nifedipine, especially in higher doses, be used with great caution, if at all. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa VERAPAMIL AND DILTIAZEM Preadministration Assessment Therapeutic Goal Verapamil and diltiazem are indicated for hypertension, angina pectoris, and cardiac dysrhythmias (atrial fibrillation, atrial flutter, and paroxysmal supraventricular tachycardia). Baseline Data For all patients, determine blood pressure and pulse rate, and obtain laboratory evaluations of liver and kidney function. For patients with angina pectoris, obtain baseline data on the frequency and severity of anginal attacks. For baseline data relevant to hypertension, see Chapter 47. Identifying High-Risk Patients Verapamil and diltiazem are contraindicated for patients with severe hypotension, sick sinus syndrome (in the absence of electronic pacing), and second-degree or third-degree AV block. Use with caution in patients with heart failure or liver impairment and in patients taking digoxin or beta blockers. Implementation: Administration Routes Oral, IV. Administration Oral. Verapamil and diltiazem may be used for angina pectoris and essential hypertension. Verapamil may be used Continued 503 Summary of Major Nursing Implicationsa—cont’d with digoxin to control ventricular rate in patients with atrial fibrillation and atrial flutter. Extended-release formulations are reserved for essential hypertension. Instruct patients to swallow extended-release formulations whole, without crushing or chewing. Before dosing, measure blood pressure and pulse rate. If hypotension or bradycardia is detected, withhold medication and notify the prescriber. Intravenous. Intravenous therapy with verapamil or diltiazem is reserved for cardiac dysrhythmias. Perform injections slowly (over 2 to 3 minutes). Monitor the ECG for AV block, sudden reduction in heart rate, and prolongation of the PR or QT interval. Have facilities for cardioversion and cardiac pacing immediately available. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Angina Pectoris. Keep an ongoing record of anginal attacks, noting the time and intensity of each attack and the likely precipitating event. Teach outpatients to chart the time, intensity, and circumstances of their attacks, and to notify the prescriber if attacks increase. Essential Hypertension. Monitor blood pressure periodically. For most patients, the goal is to reduce systolic/diastolic pressure to a value below 140/90 mm Hg. Teach patients to self-monitor their blood pressure and to maintain a blood pressure record. Minimizing Adverse Effects Cardiosuppression. Verapamil and diltiazem can cause bradycardia, AV block, and heart failure. Inform patients about manifestations of cardiac effects (e.g., slow heartbeat, shortness of breath, weight gain) and instruct them to notify the prescriber if these occur. If cardiac impairment is severe, drug use should stop. Peripheral Edema. Inform patients about signs of edema (swelling in ankles or feet), and instruct them to notify the prescriber if these occur. If necessary, edema can be reduced with a diuretic. Constipation. Constipation occurs primarily with verapamil. Advise patients that constipation can be minimized by increasing dietary fluid and fiber. Minimizing Adverse Interactions Digoxin. The combination of digoxin with verapamil or diltiazem increases the risk of partial or complete AV block. Monitor for indications of impaired AV conduction (missed beats, slowed ventricular rate). Verapamil (and possibly diltiazem) can increase plasma levels of digoxin. Digoxin dosage should be reduced. Beta Blockers. Concurrent use of a beta blocker with verapamil or diltiazem can cause bradycardia, AV block, or heart failure. Monitor closely for cardiac suppression. Administer intravenous verapamil and beta blockers several hours apart. Grapefruit Juice. Grapefruit juice can raise levels of verapamil and diltiazem. Toxicity may result. Advise patients that it may be prudent to minimize grapefruit juice consumption. Managing Acute Toxicity Remove unabsorbed drug with gastric lavage followed by activated charcoal. Give IV calcium to help counteract excessive vasodilation and reduced myocardial contractility. To raise blood pressure, give IV norepinephrine. Intravenous fluids and placing the patient in modified Trendelenburg’s position can also help. Bradycardia and AV block can be reversed with atropine and glucagon. If these are inadequate, electronic pacing may be required. DIHYDROPYRIDINES Amlodipine Clevidipine Felodipine Isradipine Nicardipine Nifedipine Nimodipine Nisoldipine Preadministration Assessment Therapeutic Goal Amlodipine, nifedipine, and nicardipine are approved for essential hypertension and angina pectoris. Isradipine, felodipine, and nisoldipine are approved for hypertension only. Nimodipine is used only for subarachnoid hemorrhage. Clevidipine is used only for IV therapy of severe hypertension. Baseline Data See nursing implications for Verapamil and Diltiazem. Identifying High-Risk Patients Use dihydropyridines with caution in patients with hypotension, sick sinus syndrome (in the absence of electronic pacing), angina pectoris (because of reflex tachycardia), heart failure, and second-degree or third-degree AV block. Implementation: Administration Route Oral. All dihydropyridines except clevidipine. Intravenous. Nicardipine, clevidipine. Administration Instruct patients to swallow sustained-release formulations whole, without crushing or chewing. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects See nursing implications for Verapamil and Diltiazem. Minimizing Adverse Effects Reflex Tachycardia. Reflex tachycardia can be suppressed with a beta blocker. Peripheral Edema. Inform patients about signs of edema and instruct them to notify the prescriber if these occur. If necessary, edema can be reduced with a diuretic. Managing Acute Toxicity Continued See nursing implications for Verapamil and Diltiazem. Patient education information is highlighted as blue text. a 504 CHAPTER 46 Preparations, Dosage, and Administration Sodium nitroprusside [Nitropress] is available in powdered form (50 mg) to be dissolved and then diluted for IV infusion. Fresh solutions may have a faint brown color. Solutions that are deeply colored (blue, green, dark red) should be discarded. Nitroprusside in solution can be degraded by light, and hence should be protected with an opaque material. Blood pressure can be adjusted to practically any level by increasing or decreasing the rate of infusion. The initial infusion rate is 0.3 mcg/kg/min. Vasodilators The maximal rate is 10 mcg/kg/min. If infusion at the maximal rate for 10 minutes fails to produce an adequate drop in blood pressure, administration should stop. During the infusion, blood pressure should be monitored continuously, with either an arterial line or an electronic monitoring device. No other drugs should be mixed with the nitroprusside solution. KEY POINTS ■ ■ ■ ■ ■ ■ Some vasodilators are selective for arterioles, some are selective for veins, and some dilate both types of vessel. Drugs that dilate arterioles reduce cardiac afterload and can thereby reduce cardiac work while increasing cardiac output and tissue perfusion. Drugs that dilate veins reduce cardiac preload and can thereby reduce cardiac work, cardiac output, and tissue perfusion. Principal indications for vasodilators are essential hypertension, hypertensive crisis, angina pectoris, heart failure, and myocardial infarction. Drugs that dilate veins can cause orthostatic hypotension. Drugs that dilate arterioles or veins can cause reflex tachycardia, which increases cardiac work and elevates blood pressure. Reflex tachycardia can be blunted with a beta blocker. ■ ■ ■ ■ ■ ■ ■ Drugs that dilate arterioles or veins can cause fluid retention, which can be blunted with a diuretic. Hydralazine causes selective dilation of arterioles. Hydralazine can cause a syndrome that resembles SLE. Minoxidil causes selective and profound dilation of arterioles. Minoxidil can cause hypertrichosis. Sodium nitroprusside dilates arterioles and veins. Prolonged infusion of nitroprusside can result in toxic accumulation of cyanide and thiocyanate. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. 509 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood BP is labetalol (20 mg by IV bolus over 2 minutes); dosing may be repeated at 10-minute intervals up to a total of 300 mg. Because severe preeclampsia can evolve into eclampsia, an antiseizure drug may be given for prophylaxis. Magnesium sulfate is the drug of choice. In one study, prophylaxis with magnesium sulfate reduced the risk of eclampsia by 58% and the risk of death by 45%. Dosing is the same as for treating eclampsia. If eclampsia develops, magnesium sulfate is the preferred drug for seizure control. Initial dosing consists of a 4- to 6-gm IV loading dose followed by 5 gm IM injected into each buttock. Maintenance consists of continuous IV infusion of 1 to 2 gm/hr or 5 gm IM injected into alternating buttocks every 4 hours. To ensure therapeutic effects and prevent toxicity, blood levels of magnesium, as well as presence of patellar reflex, should be monitored. The target range for serum magnesium is 4 to 7 mEq/L (the normal range for magnesium is 1.5 to 2 mEq/L). Can drugs help prevent preeclampsia in those at risk? Yes. When started before 16 weeks of gestation, low-dose aspirin reduces risk by about 50%. Similarly, L-arginine (combined with antioxidant vitamins) can also help. By contrast, several other preparations—magnesium, zinc, vitamin C, vitamin E, fish oil, and diuretics—appear to offer no protection at all. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Hypertension is defined as SBP greater than 130 mm Hg or DBP greater than 80 mm Hg. Primary hypertension (essential hypertension), defined as hypertension with no identifiable cause, is the most common form of hypertension. Untreated hypertension can lead to heart disease, kidney disease, and stroke. In patients older than 50, elevated systolic BP represents a greater cardiovascular risk than elevated diastolic BP. The goal of antihypertensive therapy is to decrease morbidity and mortality without decreasing quality of life. For most patients, this goal is achieved by maintaining BP between 120/80 and 130/80 mm Hg. To reduce BP, two types of treatment may be used: drug therapy and lifestyle modification (smoking cessation, reduction of salt and alcohol intake, following the DASH diet, and increasing aerobic exercise). The baroreceptor reflex, the kidneys, and the RAAS can oppose our attempts to lower BP with drugs. We can counteract the baroreceptor reflex with a beta blocker, the kidneys with a diuretic, and the RAAS with an ACE inhibitor, ARB, DRI, or aldosterone antagonist. Thiazide diuretics (e.g., hydrochlorothiazide, chlorthalidone) and loop diuretics (e.g., furosemide) reduce BP in two ways: they reduce blood volume (by promoting diuresis) and they reduce arterial resistance (by an unknown mechanism). Loop diuretics should be reserved for (1) patients who need greater diuresis than can be achieved with thiazides and (2) patients with a low GFR (because thiazides don’t work when GFR is low). Beta blockers (e.g., metoprolol) appear to lower BP primarily by reducing peripheral vascular resistance; the mechanism is unknown. They may also lower BP by decreasing myocardial contractility and suppressing reflex tachycardia (through beta1 blockade in the heart), and by decreasing renin release (through beta1 blockade in the kidney). ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Calcium channel blockers (e.g., diltiazem, nifedipine) reduce BP by promoting dilation of arterioles. ACE inhibitors, ARBs, and DRIs lower BP by preventing angiotensin II–mediated vasoconstriction and aldosteronemediated volume expansion. ACE inhibitors work by blocking the formation of angiotensin II, whereas ARBs block the actions of angiotensin II. DRIs prevent formation of angiotensin I and thereby shut down the entire RAAS. Aldosterone antagonists lower BP by preventing aldosteronemediated retention of sodium and water in the kidney. Thiazide diuretics are preferred drugs for initial therapy of uncomplicated hypertension. When a combination of drugs is used for hypertension, each drug should have a different mechanism of action. Dosages of antihypertensive drugs should be low initially and increased gradually. This approach minimizes adverse effects and permits baroreceptors to reset to a lower pressure. Lack of patient adherence is the major cause of treatment failure in antihypertensive therapy. Adherence is difficult to achieve because (1) hypertension has no symptoms (so drug benefits aren’t obvious); (2) hypertension progresses slowly (so patients think they can postpone treatment); and (3) treatment is complex and expensive, continues lifelong, and can cause adverse effects. A severe hypertensive emergency exists when diastolic BP exceeds 120 mm Hg and there is ongoing end-organ damage. Nitroprusside (IV) is a drug of choice for hypertensive emergencies. Hypertension is the most common complication of pregnancy. Methyldopa and labetalol are drugs of choice for treating chronic hypertension of pregnancy. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. 526 CHAPTER 47 Drugs for Hypertension Summary of Major Nursing Implicationsa ANTIHYPERTENSIVE DRUGS Preadministration Assessment Therapeutic Goal The goal of antihypertensive therapy is to prevent the longterm sequelae of hypertension (heart disease, kidney disease, stroke) while minimizing drug effects that can reduce quality of life. For most patients, BP should be reduced to less than 130/80 mm Hg. Baseline Data The following tests should be done in all patients: BP; electrocardiogram; complete urinalysis; hemoglobin and hematocrit; and blood levels of sodium, potassium, calcium, creatinine, glucose, uric acid, triglycerides, and cholesterol (total, LDL, and HDL cholesterol). Identifying High-Risk Patients When taking the patient’s drug history, attempt to identify drugs that can raise BP or that can interfere with the effects of antihypertensive drugs. Some drugs of concern are listed later under Minimizing Adverse Interactions. The patient history should identify comorbid conditions that either contraindicate the use of specific agents (e.g., AV block contraindicates use of beta blockers) or require that drugs be used with special caution (e.g., thiazide diuretics must be used with caution in patients with gout or diabetes). For risk factors that pertain to specific antihypertensive drugs, see the chapters in which those drugs are discussed. Implementation: Administration Routes All drugs for chronic hypertension are administered orally. Dosage To minimize adverse effects, dosages should be low initially and increased gradually. It is counterproductive to employ high initial dosages that produce a rapid fall in pressure while also producing intense adverse effects that can discourage adherence. After 12 months of successful treatment, dosage reductions should be tried. Implementation: Measures to Enhance Therapeutic Effects Lifestyle Modifications In hypertensive patients, lifestyle changes can reduce BP and increase responsiveness to antihypertensive drugs. These changes should be tried for 6 to 12 months before implementing drug therapy and should continue even if drugs are required. Weight Reduction. Help patients develop an exercise and weight management program if needed. Sodium Restriction. Encourage patients to consume no more than 2300 mg of salt daily and provide them with information on the salt content of foods. DASH Diet. Encourage patients to adopt a diet rich in fruits, vegetables, and low-fat dairy products, and low in total fat, unsaturated fat, and cholesterol. Alcohol Restriction. Encourage patients to limit alcohol consumption to 1 ounce/day (for most men) and 0.5 ounce/ day (for women and lighter weight men). One ounce of ethanol is equivalent to about two mixed drinks, two glasses of wine, or two cans of beer. Exercise. Encourage patients with a sedentary lifestyle to perform 30 to 45 minutes of aerobic exercise (e.g., walking, swimming, bicycling) most days of the week. Smoking Cessation. Strongly encourage patients to quit smoking. Teach patients about aids for smoking cessation (e.g., nicotine patch, bupropion, varenicline). Promoting Adherence Nonadherence is the major cause of treatment failure. Achieving adherence is difficult for several reasons: hypertension is devoid of overt symptoms; drugs don’t make people feel better (but can make them feel worse); regimens can be complex and expensive; complications of hypertension take years to develop, thereby providing a misguided rationale for postponing treatment; and treatment usually lasts lifelong. Provide Patient Education. Educate patients about the long-term consequences of hypertension and the ability of lifestyle changes and drug therapy to decrease morbidity and prolong life. Inform patients that drugs do not cure hypertension; therefore, the medication prescribed must usually be taken lifelong. Encourage Self-Monitoring. Make certain that patients know the treatment goal (usually reduction of BP to less than 130/80 mm Hg), and teach them to monitor and chart their own BP. This will increase their involvement and help them see the benefits of treatment. Minimize Adverse Effects. Adverse drug effects are an obvious deterrent to adherence. Measures to reduce undesired effects are discussed under Minimizing Adverse Effects. Establish a Collaborative Relationship. Encourage patients to be active partners in setting treatment goals, creating a treatment program, and evaluating progress. Simplify the Regimen. An antihypertensive regimen can consist of several drugs taken multiple times a day. Once an effective regimen has been established, attempt to switch to once-a-day or twice-a-day dosing. If an appropriate combination product is available (e.g., a fixed-dose combination of a thiazide diuretic plus an ACE inhibitor), substitute the combination product for its components. Other Measures. Additional measures to promote adherence include providing positive reinforcement when treatment goals are achieved, involving family members in the treatment program, scheduling office visits at convenient times, following up on patients who miss an appointment, and devising a program that is effective but keeps costs low. Continued 527 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d Ongoing Evaluation and Interventions Evaluating Treatment Monitor BP periodically. The usual goal is to reduce it to less than 130/80 mm Hg. Teach patients to self-monitor their BP and to maintain a BP record. Minimizing Adverse Effects General Considerations. The fundamental strategy for decreasing adverse effects is to tailor the regimen to the sensitivities of the patient. If a drug causes objectionable effects, a more acceptable drug should be substituted. Inform patients about the potential side effects of treatment, and encourage them to report objectionable responses. Avoid drugs that can exacerbate comorbid conditions. For example, don’t give beta blockers to patients who have bradycardia, AV block, or asthma. Table 47.5 lists drugs to avoid in patients with specific disorders. Initiate therapy with low doses and increase them gradually. Adverse Effects of Specific Drugs. For measures to minimize adverse effects of specific antihypertensive drugs (e.g., beta blockers, diuretics, ACE inhibitors), see the chapters in which those drugs are discussed. Minimizing Adverse Interactions When taking the patient history, identify drugs that can raise BP or interfere with the effects of antihypertensive drugs. Drugs of concern include oral contraceptives, nonsteroidal anti-inflammatory drugs, glucocorticoids, appetite suppressants, tricyclic antidepressants, monoamine oxidase inhibitors, cyclosporine, erythropoietin, alcohol (in large quantities), and nasal decongestants and other cold remedies. Antihypertensive regimens frequently contain two or more drugs, posing a potential risk of adverse interactions (e.g., ACE inhibitors can increase the risk of hyperkalemia caused by potassium-sparing diuretics). For interactions that pertain to specific antihypertensive drugs, see the chapters in which those drugs are discussed. Patient education information is highlighted as blue text. a 528 CHAPTER 48 Device Therapy Implanted Cardioverter-Defibrillators. Cardiac arrest and fatal ventricular dysrhythmias are relatively common complications of HF. Accordingly, implantable cardioverterdefibrillators are now recommended for primary or secondary prevention to reduce mortality in selected patients. Cardiac Resynchronization. When the left and right ventricles fail to contract at the same time, cardiac output is further compromised. Synchronized contractions can be restored with a biventricular pacemaker. In clinical trials, cardiac resynchronization improved exercise tolerance and quality of life and reduced all-cause mortality. Exercise Training In the past, bed rest was recommended owing to concern that physical activity might accelerate progression of LV dysfunction. However, we now know that inactivity is actually detrimental: It reduces conditioning, worsens exercise intolerance, and contributes to HF symptoms. Conversely, studies have shown that exercise training can improve clinical status, increase exercise capacity, and improve quality of life. Accordingly, exercise training should be considered for all stable patients. Evaluating Treatment Evaluation is based on symptoms and physical findings. Reductions in dyspnea on exertion, paroxysmal nocturnal dyspnea, and orthopnea (difficulty breathing, except in the upright position) indicate success. The physical examination should assess for reductions in jugular distention, edema, and crackles. Success is also indicated by increased capacity for physical activity. Accordingly, patients should be interviewed to determine improvements in the maximal activity they can perform without symptoms, the type of activity that regularly produces symptoms, and the maximal activity they can tolerate. (Activity is defined as walking, stair climbing, activities of daily living, or any other activity that is appropriate.) Successful treatment should also improve health-related quality of life in general. Thus the interview should look for improvements in sleep, sexual function, outlook on life, cognitive function (alertness, Drugs for Heart Failure memory, concentration), and ability to participate in usual social, recreational, and work activities. Routine measurement of ejection fraction or maximal exercise capacity is not recommended. Although the degree of reduction in ejection fraction measured at the beginning of therapy is predictive of outcome, improvement in the ejection fraction does not necessarily indicate the prognosis has changed. As noted earlier, a reduction in circulating BNP indicates improvement. The lower BNP is, the better the odds of long-term survival. Stage D Patients in Stage D have advanced structural heart disease and marked symptoms of HF at rest, despite treatment with maximal dosages of medications used in Stage C. Repeated and prolonged hospitalization is common. For eligible candidates, the best long-term solution is a heart transplant. An implantable LV mechanical assist device can be used as a “bridge” in patients awaiting a transplant and to prolong life in those who are not transplant eligible. Management focuses largely on the control of fluid retention, which underlies most signs and symptoms. Intake and output should be monitored closely, and the patient should be weighed daily. Fluid retention can usually be treated with a loop diuretic, perhaps combined with a thiazide. If volume overload becomes severe, the patient should be hospitalized and given an IV diuretic. If needed, IV dopamine or IV dobutamine can be added to increase renal blood flow, thereby enhancing diuresis. Patients should not be discharged until a stable and effective oral diuretic regimen has been established. What about beta blockers and ACE inhibitors? These agents may be tried, but doses should be low and responses monitored with care. In Stage D, beta blockers pose a significant risk of making HF worse, and ACE inhibitors may induce profound hypotension or renal failure. When severe symptoms persist despite application of all recommended therapies, options for end-of-life care should be discussed with the patient and family. KEY POINTS ■ ■ ■ Heart failure with LV systolic dysfunction, referred to simply as heart failure (HF) in this chapter, is characterized by ventricular dysfunction, reduced cardiac output, signs of inadequate tissue perfusion (fatigue, shortness of breath, exercise intolerance), and signs of fluid overload (venous distention, peripheral edema, pulmonary edema). The initial phase of HF consists of cardiac remodeling—a process in which the ventricles dilate (grow larger), hypertrophy (increase in wall thickness), and become more spherical—coupled with cardiac fibrosis and myocyte death. As a result of these changes, cardiac output is reduced. Reduced cardiac output leads to compensatory responses: (1) activation of the SNS, (2) activation of the RAAS, and (3) retention of water and expansion of blood volume. As a result of volume expansion, cardiac dilation increases. ■ ■ ■ ■ If the compensatory responses are not sufficient to maintain adequate production of urine, body water will continue to accumulate, eventually causing death (from complete cardiac failure secondary to excessive cardiac dilation and cardiac edema). There are three major groups of drugs for heart failure: diuretics, ACE inhibitors or ARBs, and beta blockers. Digoxin, which had been used widely in the past, may be added as indicated. Diuretics are first-line drugs for all patients with fluid overload. By reducing blood volume, these drugs can decrease venous pressure, arterial pressure, pulmonary edema, peripheral edema, and cardiac dilation. Although diuretics can reduce symptoms of HF, they do not prolong survival. Continued 543 UNIT VII ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Drugs That Affect the Heart, Blood Vessels, and Blood Thiazide diuretics are ineffective when GFR is low, and cannot be used if cardiac output is greatly reduced. Loop diuretics are effective even when GFR is low, and are preferred to thiazides for most patients. Thiazide diuretics and loop diuretics can cause hypokalemia and can increase the risk of digoxin-induced dysrhythmias. Potassium-sparing diuretics are used to counteract potassium loss caused by thiazide diuretics and loop diuretics. Potassium-sparing diuretics can cause hyperkalemia. By doing so, they can increase the risk of hyperkalemia in patients taking ACE inhibitors or ARBs. In patients with HF, ACE inhibitors improve functional status and reduce mortality. In the absence of specific contraindications, all patients should be prescribed one. ACE inhibitors block formation of angiotensin II, promote accumulation of kinins, and reduce aldosterone release. As a result, these drugs cause dilation of veins and arterioles, promote renal excretion of water, and favorably alter cardiac remodeling. By dilating arterioles, ACE inhibitors (1) improve regional blood flow in the kidneys and other tissues and (2) reduce cardiac afterload, which causes stroke volume and cardiac output to rise. By dilating veins, ACE inhibitors reduce venous pressure, which in turn reduces pulmonary congestion, peripheral edema, preload, and cardiac dilation. By suppressing aldosterone release, ACE inhibitors increase excretion of sodium and water, and decrease excretion of potassium. By increasing levels of kinins (and partly by decreasing levels of angiotensin II), ACE inhibitors can favorably alter cardiac remodeling. Major side effects of ACE inhibitors are hypotension, hyperkalemia, cough, angioedema, and birth defects. ARBs share the beneficial hemodynamic effects of ACE inhibitors, but not the beneficial effects on cardiac remodeling. In patients with HF, ARBs should be reserved for patients intolerant of ACE inhibitors (usually owing to cough). In patients with HF, aldosterone antagonists (e.g., spironolactone, eplerenone) reduce symptoms and prolong life. Benefits derive from blocking aldosterone receptors in the heart and blood vessels. Beta blockers can prolong survival in patients with HF, and are considered first-line therapy. To avoid excessive cardiosuppression, beta-blocker dosage must be very low initially and then gradually increased. Isosorbide dinitrate (which dilates veins) plus hydralazine (which dilates arterioles) can be used in place of an ACE inhibitor (or ARB) if an ACE inhibitor (or ARB) cannot be used. BiDil, a fixed-dose combination of hydralazine and isosorbide dinitrate, is approved specifically for treating HF in African Americans. Digoxin and other inotropic agents increase the force of myocardial contraction and thereby increase cardiac output. Of the available inotropic agents, digoxin is the only one that is both effective and safe when used orally and the only one suitable for long-term use. ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Digoxin increases contractility by inhibiting myocardial Na+/K+-ATPase, thereby (indirectly) increasing intracellular calcium, which in turn facilitates the interaction of actin and myosin. Potassium competes with digoxin for binding to Na+/K+ATPase. Therefore, if potassium levels are low, excessive inhibition of Na+/K+-ATPase can occur, resulting in toxicity. Conversely, if potassium levels are high, insufficient inhibition can occur, resulting in therapeutic failure. Accordingly, it is imperative to keep potassium levels in the normal physiologic range: 3.5 to 5 mEq/L. By increasing cardiac output, digoxin can reverse all of the overt manifestations of HF: cardiac output improves, heart rate decreases, heart size declines, constriction of arterioles and veins decreases, water retention reverses, blood volume declines, peripheral and pulmonary edema decrease, water weight is lost, and exercise tolerance improves. Unfortunately, although digoxin can improve symptoms, it does not prolong life. In patients with HF, benefits of digoxin are not due solely to improved cardiac output; neurohormonal effects are important too. Digoxin causes dysrhythmias by altering the electrical properties of the heart (secondary to inhibition of Na+/K+ATPase). The most common reason for digoxin-related dysrhythmias is diuretic-induced hypokalemia. If a severe digoxin overdose is responsible for dysrhythmias, digoxin levels can be lowered using Fab antibody fragments [Digifab]. In addition to dysrhythmias, digoxin can cause GI effects (anorexia, nausea, vomiting) and CNS effects (fatigue, visual disturbances). Gastrointestinal and CNS effects often precede dysrhythmias and therefore can provide advance warning of serious toxicity. Digoxin has a narrow therapeutic range. Digoxin is eliminated by renal excretion. Although routine monitoring of digoxin levels is generally unnecessary, monitoring can be helpful when dosage is changed, symptoms of HF intensify, kidney function declines, signs of toxicity appear, or drugs that affect digoxin levels are added to or deleted from the regimen. Maintenance doses of digoxin are based primarily on observation of the patient: Doses should be large enough to minimize symptoms of HF but not so large as to cause adverse effects. Maintenance doses of digoxin must be reduced if renal function declines. Therapy of Stage C HF has four major goals: (1) relief of pulmonary and peripheral congestion, (2) improvement of functional status and quality of life, (3) delay of progression of cardiac remodeling and LV dysfunction, and (4) prolongation of life. For routine therapy, Stage C HF is treated with a diuretic, an ACE inhibitor or an ARB, and a beta blocker. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. 544 Summary of Major Nursing Implicationsa DIGOXIN Preadministration Assessment Therapeutic Goal Digoxin is used to treat HF and dysrhythmias. Be sure to confirm which disorder the drug has been ordered for. Baseline Data Assess for signs and symptoms of HF, including fatigue, weakness, cough, breathing difficulty (orthopnea, dyspnea on exertion, paroxysmal nocturnal dyspnea), jugular distention, and edema. Determine baseline values for maximal activity without symptoms, activity that regularly causes symptoms, and maximal tolerated activity. Laboratory tests should include an ECG, serum electrolytes, measurement of ejection fraction, and evaluation of kidney function. Identifying High-Risk Patients Digoxin is contraindicated for patients experiencing ventricular fibrillation, ventricular tachycardia, or digoxin toxicity. Exercise caution in the presence of conditions that can predispose the patient to serious adverse responses to digoxin, such as hypokalemia, partial AV block, advanced HF, or renal impairment. Implementation: Administration Routes Oral, slow IV injection. Administration Oral. Determine heart rate and rhythm before administration. If heart rate is less than 60 beats/min or if a change in rhythm is detected, withhold digoxin and notify the prescriber. Warn patients not to “double up” on doses in attempts to compensate for missed doses. Intravenous. Monitor cardiac status closely for 1 to 2 hours following IV injection. Promoting Adherence Because digoxin has a narrow therapeutic range, rigid adherence to the prescribed dosage is essential. Inform patients that failure to take digoxin exactly as prescribed may lead to toxicity or therapeutic failure. If poor adherence is sus- pected, serum drug levels may help in assessing the extent of nonadherence. Implementation: Measures to Enhance Therapeutic Effects Advise patients to limit salt intake to 1500 mg/day and to avoid excessive fluids. Advise patients who drink alcohol to consume no more than one drink each day. Help patients establish an appropriate program of regular mild exercise (e.g., walking, cycling). Precipitating factors for HF (e.g., hypertension, valvular heart disease) should be corrected. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Evaluation is based on symptoms and physical findings. Assess for reductions in orthopnea, dyspnea on exertion, paroxysmal nocturnal dyspnea, neck vein distention, edema, and crackles, and for increased capacity for physical activity. In addition, Patient education information is highlighted as blue text. a assess for improvements in sleep, sexual function, outlook on life, cognitive function, and ability to participate in social, recreational, and work activities. Plasma BNP levels reflect cardiac status: The lower the level, the better the odds for long-term survival. Measurement of plasma drug levels can help determine the cause of therapeutic failure. The optimal range for digoxin is 0.5 to 0.8 ng/mL. Minimizing Adverse Effects Cardiotoxicity. Dysrhythmias are the most serious adverse effect of digoxin. Monitor hospitalized patients for alterations in heart rate or rhythm, and withhold digoxin if significant changes develop. Inform outpatients about the danger of dysrhythmias. Teach them to monitor their pulses for rate and rhythm, and instruct them to notify the prescriber if significant changes occur. Provide the patient with an ECG rhythm strip; this can be used by providers unfamiliar with the patient (e.g., when the patient is traveling) to verify suspected changes in rhythm. Hypokalemia, usually diuretic induced, is the most frequent underlying cause of dysrhythmias. Monitor serum potassium concentrations. If hypokalemia develops, potassium levels can be raised with potassium supplements, a potassium-sparing diuretic, or both. Teach patients to recognize early signs of hypokalemia (e.g., muscle weakness), and instruct them to notify the prescriber if these develop. Severe vomiting and diarrhea can increase potassium loss; exercise caution if these events occur. To treat digoxin-induced dysrhythmias: (1) withdraw digoxin and diuretics (make sure that a written order for digoxin withdrawal is made); (2) administer potassium (unless potassium levels are above normal or AV block is present); (3) administer an antidysrhythmic drug (phenytoin or lidocaine, but not quinidine) if indicated; (4) manage bradycardia with atropine or electrical pacing; and (5) treat with Fab fragments if toxicity is life threatening. Noncardiac Effects. Nausea, vomiting, anorexia, fatigue, and visual disturbances (blurred or yellow vision) frequently foreshadow more serious toxicity (dysrhythmias) and should be reported immediately. Inform patients about these early indications of toxicity, and instruct them to notify the prescriber if they develop. Minimizing Adverse Interactions Diuretics. Thiazide diuretics and loop diuretics increase the risk of dysrhythmias by promoting potassium loss. Monitor potassium levels. If hypokalemia develops, it should be corrected with potassium supplements, a potassium-sparing diuretic, or both. ACE Inhibitors and ARBs. These drugs can elevate potassium levels and decrease therapeutic responses to digoxin. Exercise caution if an ACE inhibitor or ARB is combined with potassium supplements or a potassium-sparing diuretic. Sympathomimetic Agents. Sympathomimetic drugs (e.g., dopamine, dobutamine) stimulate the heart, thereby increasing the risk of tachydysrhythmias and ectopic pacemaker activity. When sympathomimetics are combined with digoxin, monitor closely for dysrhythmias. Quinidine. Quinidine can elevate plasma levels of digoxin. If quinidine is employed concurrently with digoxin, digoxin dosage must be reduced. Do not use quinidine to treat digoxininduced dysrhythmias. UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Dysrhythmias result from alteration of the electrical impulses that regulate cardiac rhythm. Antidysrhythmic drugs control rhythm by correcting or compensating for these alterations. In the healthy heart, the SA node is the pacemaker. Impulses originating in the SA node must travel through the AV node to reach the ventricles. Impulses arriving at the AV node are delayed before going on to excite the ventricles. The His-Purkinje system conducts impulses rapidly throughout the ventricles, thereby causing all parts of the ventricles to contract in near synchrony. The heart employs two kinds of action potentials: fast potentials and slow potentials. Fast potentials occur in the His-Purkinje system, atrial muscle, and ventricular muscle. Slow potentials occur in the SA node and AV node. Phase 0 of fast potentials (depolarization) is generated by rapid influx of sodium. Because depolarization is fast, these potentials conduct rapidly. During phase 2 of fast potentials, calcium enters myocardial cells, thereby promoting contraction. Phase 3 of fast potentials (repolarization) is generated by rapid extrusion of potassium. Phase 0 of slow potentials (depolarization) is caused by slow influx of calcium. Because depolarization is slow, these potentials conduct slowly. Spontaneous phase 4 depolarization—of fast or slow potentials—gives cells automaticity. Spontaneous phase 4 depolarization of cells in the SA node normally determines heart rate. The P wave of an ECG is caused by depolarization of the atria. The QRS complex is caused by depolarization of the ventricles. Widening of the QRS complex indicates slowed conduction through the ventricles. The T wave is caused by repolarization of the ventricles. The PR interval represents the time between onset of the P wave and onset of the QRS complex. PR prolongation indicates delayed AV conduction. The QT interval represents the time between onset of the QRS complex and completion of the T wave. QT prolongation indicates delayed ventricular repolarization. Dysrhythmias arise from disturbances of impulse formation (automaticity) or impulse conduction. Reentrant dysrhythmias result from a localized, selfsustaining circuit capable of repetitive cardiac stimulation. Tachydysrhythmias can be divided into two major groups: supraventricular tachydysrhythmias and ventricular tachydysrhythmias. In general, ventricular tachydysrhythmias disrupt cardiac pumping more than do supraventricular tachydysrhythmias. Treatment of supraventricular tachydysrhythmias is often directed at blocking impulse conduction through the AV node, rather than at eliminating the dysrhythmia. Treatment of ventricular dysrhythmias is usually directed at eliminating the dysrhythmia. ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ 564 All antidysrhythmic drugs are also prodysrhythmic (proarrhythmic). That is, they all can worsen existing dysrhythmias and generate new ones. Class I antidysrhythmic drugs block cardiac sodium channels and thereby slow impulse conduction through the atria, ventricles, and His-Purkinje system. Slowing ventricular conduction widens the QRS complex. Quinidine (a class IA drug) blocks sodium channels and delays ventricular repolarization. Delaying ventricular repolarization prolongs the QT interval. Quinidine causes diarrhea and other GI symptoms in 33% of patients. These effects frequently force drug withdrawal. Quinidine can cause dysrhythmias. Widening of the QRS complex (by 50% or more) and excessive prolongation of the QT interval are warning signs. Quinidine can raise digoxin levels. If the drugs are used together, digoxin dosage must be reduced. Class IB agents differ from class IA agents in two ways: they accelerate repolarization and have little or no effect on the ECG. Lidocaine (a class IB agent) is used only for ventricular dysrhythmias. The drug is not active against supraventricular dysrhythmias. Lidocaine undergoes rapid inactivation by the liver. As a result, it must be administered by continuous IV infusion. Propranolol and other class II drugs block cardiac beta1 receptors. By blocking cardiac beta1 receptors, propranolol attenuates sympathetic stimulation of the heart and thereby decreases SA nodal automaticity, AV conduction velocity, and myocardial contractility. By decreasing AV conduction velocity, propranolol prolongs the PR interval. The effects of propranolol on the heart result (ultimately) from suppressing calcium entry. Therefore, the cardiac effects of propranolol and the effects of calcium channel blockers are nearly identical. Propranolol is especially useful for treating dysrhythmias caused by excessive sympathetic stimulation of the heart. In patients with supraventricular tachydysrhythmias, propranolol helps by (1) slowing discharge of the SA node and (2) decreasing impulse conduction through the AV node, which prevents the atria from driving the ventricles at an excessive rate. Class III antidysrhythmics block potassium channels and thereby delay repolarization of fast potentials. As a result, they prolong the action potential duration and the effective refractory period. By delaying ventricular repolarization, they prolong the QT interval. Amiodarone (a class III agent) is highly effective against atrial and ventricular dysrhythmias, but can cause multiple serious adverse effects, including damage to the lungs, eyes, liver, and thyroid. Dronedarone, a derivative of amiodarone, is somewhat less toxic than amiodarone, but also less effective. In patients with heart failure or permanent atrial fibrillation, dronedarone doubles the risk of death. CHAPTER 49 ■ ■ ■ Verapamil and diltiazem (class IV antidysrhythmics) block cardiac calcium channels and thereby reduce automaticity of the SA node, slow conduction through the AV node, and decrease myocardial contractility. These effects are identical to those of the beta blockers. By suppressing AV conduction, verapamil and diltiazem prolong the PR interval. Verapamil and diltiazem are used to slow ventricular rate in patients with atrial fibrillation or atrial flutter and to terminate SVT caused by an AV nodal reentrant circuit. ■ ■ Antidysrhythmic Drugs In both cases, benefits derive from suppressing AV nodal conduction. Adenosine is a drug of choice for terminating paroxysmal SVT. Adenosine has a very short half-life (less than 10 seconds) and must be given by IV bolus. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa Summaries are limited to the major antidysrhythmic drugs. Summaries for beta blockers (propranolol, acebutolol, and esmolol), phenytoin, calcium channel blockers (verapamil and diltiazem), and digoxin appear in Chapters 18, 24, 45, and 48, respectively. QUINIDINE Preadministration Assessment Therapeutic Goal The usual goal is long-term suppression of atrial and ventricular dysrhythmias. Baseline Data Obtain a baseline ECG and laboratory evaluation of liver function. Determine blood pressure. Identifying High-Risk Patients Quinidine is contraindicated for patients with a history of hypersensitivity to quinidine or other cinchona alkaloids and for patients with complete heart block, digoxin toxicity, or conduction disturbances associated with marked QRS widening and QT prolongation. Exercise caution in patients with partial AV block, HF, hypotensive states, and hepatic dysfunction. Minimizing Adverse Effects Diarrhea. Diarrhea and other GI disturbances occur in one-third of patients and frequently force drug withdrawal. Inform patients that they can reduce GI effects by taking quinidine with meals. Cinchonism. Inform patients about symptoms of cinchonism (tinnitus, headache, nausea, vertigo, disturbed vision), and instruct them to notify the prescriber if these develop. Cardiotoxicity. Monitor the ECG for signs of cardiotoxicity, especially widening of the QRS complex (by 50% or more) and excessive prolongation of the QT interval. Monitor pulses for significant changes in rate or regularity. If signs of cardiotoxicity develop, withhold quinidine and notify the prescriber. Arterial Embolism. Embolism may occur during therapy of atrial fibrillation. Risk is reduced by treatment with an anticoagulant (e.g., warfarin, dabigatran). Observe for signs of thromboembolism (e.g., sudden chest pain, dyspnea), and report these immediately. Minimizing Adverse Interactions Digoxin. Quinidine can double digoxin levels. When these drugs are combined, digoxin dosage should be reduced. Monitor patients for digoxin toxicity (dysrhythmias). PROCAINAMIDE Implementation: Administration Routes Usual Route. Oral. Rare Routes. IM and IV. Administration Advise patients to take quinidine with meals. Warn them not to crush or chew sustained-release formulations. Dosage size depends on the particular quinidine salt being used: 200 mg of quinidine sulfate is equivalent to 275 mg of quinidine gluconate. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Monitor for beneficial changes in the ECG. Plasma drug levels should be kept between 2 and 5 mcg/mL. Preadministration Assessment Therapeutic Goal Procainamide is indicated for acute and long-term management of ventricular and supraventricular dysrhythmias. Because procainamide can be toxic with long-term use, quinidine is preferred to procainamide for chronic suppression. Baseline Data Obtain a baseline ECG, complete blood count, and laboratory evaluations of liver and kidney function. Determine blood pressure. Identifying High-Risk Patients Procainamide is contraindicated for patients with systemic lupus erythematosus (SLE), complete AV block, and secondContinued 565 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d degree or third-degree AV block in the absence of an electronic pacemaker, and patients with a history of procaine allergy. Exercise caution in patients with hepatic or renal dysfunction. LIDOCAINE Implementation: Administration Routes Oral, IM, IV. Baseline Data Obtain a baseline ECG and determine blood pressure. Administration Instruct patients to administer procainamide at evenly spaced intervals around-the-clock. Warn patients not to crush or chew sustained-release formulations. When switching from IV procainamide to oral procainamide, allow 3 hours to elapse between stopping the infusion and giving the first oral dose. Give IM injections deep into the gluteal muscle. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Monitor the ECG for beneficial changes. Plasma drug levels should be kept between 3 and 10 mcg/mL. Minimizing Adverse Effects SLE-like Syndrome. Prolonged therapy can produce a syndrome resembling SLE. Inform patients about manifesta- tions of SLE (joint pain and inflammation; hepatomegaly; unexplained fever; soreness of the mouth, throat, or gums), and instruct them to notify the prescriber if these develop. If SLE is diagnosed, procainamide should be discontinued. If discontinuation is impossible, signs and symptoms can be controlled with a nonsteroidal anti-inflammatory drug (e.g., aspirin) or a glucocorticoid. The ANA titer should be measured periodically, and if it rises, procainamide withdrawal should be considered. Blood Dyscrasias. Procainamide can cause agranulocytosis, thrombocytopenia, and neutropenia. Deaths have occurred. Obtain complete blood counts weekly during the first 3 months of treatment and periodically thereafter. Instruct patients to inform the prescriber at the first sign of infection (fever, chills, sore throat), bruising, or bleeding. If subsequent blood counts indicate hematologic disturbance, discontinue procainamide immediately. Cardiotoxicity. Procainamide can cause dysrhythmias. Monitor pulses for changes in rate or regularity. Monitor the ECG for excessive QRS widening (greater than 50%) and for PR prolongation. If these occur, withhold procainamide and notify the prescriber. Arterial Embolism. Embolism may occur during therapy of atrial fibrillation. Risk is reduced by treatment with an anticoagulant (e.g., warfarin, dabigatran). Observe for signs of thromboembolism (e.g., sudden chest pain, dyspnea), and report these immediately. Preadministration Assessment Therapeutic Goal Acute management of ventricular dysrhythmias. Identifying High-Risk Patients Lidocaine is contraindicated for patients with Stokes-Adams syndrome, Wolff-Parkinson-White syndrome, and severe degrees of SA, AV, or intraventricular block in the absence of electronic pacing. Exercise caution in patients with hepatic dysfunction or impaired hepatic blood flow. Implementation: Administration Routes Usual. IV. Emergencies. IM. Administration Intravenous. Make certain the lidocaine preparation is labeled for IV use (i.e., is devoid of preservatives and catecholamines). Dilute concentrated preparations with 5% dextrose in water. The initial dose is 50 to 100 mg (1 mg/kg) infused at a rate of 25 to 50 mg/min. For maintenance, monitor the ECG and adjust the infusion rate on the basis of cardiac response. The usual rate is 1 to 4 mg/min. Intramuscular. Reserve for emergencies. The usual dose is 300 mg injected into the deltoid muscle. Switch to IV lidocaine as soon as possible. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Continuous ECG monitoring is required. Plasma drug levels should be kept between 1.5 and 5 mcg/mL. Minimizing Adverse Effects Excessive doses can cause convulsions and respiratory arrest. Equipment for resuscitation should be available. Seizures can be managed with diazepam. AMIODARONE Preadministration Assessment Therapeutic Goal Oral Therapy. Long-term treatment of (1) atrial fibrillation and (2) life-threatening recurrent ventricular fibrillation or recurrent hemodynamically unstable ventricular tachycardia in patients who have not responded to safer drugs. Intravenous Therapy. Initial treatment of recurrent ventricular fibrillation, shock-resistant ventricular fibrillation, 566 CHAPTER 49 Antidysrhythmic Drugs Summary of Major Nursing Implicationsa—cont’d recurrent hemodynamically unstable ventricular tachycardia, atrial fibrillation, and AV nodal reentrant tachycardia. Baseline Data Obtain a baseline ECG, eye examination, and chest x-ray, along with potassium and magnesium levels, and tests for thyroid, pulmonary, and liver function. Identifying High-Risk Patients Amiodarone is contraindicated for patients with severe sinus node dysfunction or second- or third-degree AV block, and for women who are pregnant or breast-feeding. Exercise caution in patients with thyroid disorders, hypokalemia, or hypomagnesemia. Implementation: Administration Routes Oral. Used for maintenance therapy of atrial and ventricular dysrhythmias. Intravenous. Used for acute therapy of atrial and ventricular dysrhythmias. Administration and Dosage Oral. Initiate treatment in a hospital. High doses are used initially (800 to 1600 mg/day for 1 to 3 weeks). The usual maintenance dosage is 400 mg/day. Intravenous. Administer by continuous IV infusion, starting with a rapid infusion rate and later reducing the rate for maintenance. Intravenous treatment may last from 2 days to 3 weeks. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Monitor for beneficial changes in the ECG. Minimizing Adverse Effects Pulmonary Toxicity. Amiodarone can cause potentially fatal lung damage (hypersensitivity pneumonitis, interstitial/ alveolar pneumonitis, and pulmonary fibrosis). Obtain a baseline chest x-ray and pulmonary function test, and monitor pulmonary function throughout treatment. Inform patients about signs of lung injury (dyspnea, cough, chest pain), and instruct them to report these immediately. Treatment consists of withdrawing amiodarone and providing supportive care, sometimes including glucocorticoids. Cardiotoxicity. Amiodarone can cause HF and atrial and ventricular dysrhythmias. Patients with pre-existing heart failure must not use the drug. Warn patients about signs of HF (e.g., shortness of breath, reduced exercise tolerance, fatigue, tachycardia, weight gain), and instruct them to report these immediately. Liver Toxicity. Amiodarone can injure the liver. Obtain tests of liver function at baseline and periodically during treatment. If circulating liver enzymes exceed 3 times the normal level, amiodarone should be withdrawn. Inform patients about signs and symptoms of liver injury (e.g., anorexia, nausea, vomiting, malaise, fatigue, itching, jaundice, dark urine), and instruct them to report them immediately. Thyroid Toxicity. Amiodarone can cause hypothyroidism and hyperthyroidism. Obtain tests of thyroid function at baseline and periodically during treatment. Treat hypothyroidism with thyroid hormone supplements. Treat hyperthyroidism with an antithyroid drug (e.g., methimazole) or thyroidectomy. Stopping amiodarone should be considered. Toxicity in Pregnancy and Breast-Feeding. Amiodarone can harm the developing fetus and breast-feeding infant. Warn patients to avoid pregnancy and breast-feeding while using amiodarone and for several months after stopping. Ophthalmic Effects. Amiodarone has been associated with optic neuropathy and optic neuritis, sometimes progressing to blindness. Obtain ophthalmic tests, including funduscopy and a slit-lamp examination, at baseline and periodically during treatment. Advise patients to report reductions in visual acuity or peripheral vision. If optic neuropathy or neuritis is diagnosed, discontinuing amiodarone should be considered. Virtually all patients develop corneal microdeposits. In most cases, the deposits have no effect on vision and thus only rarely lead to the discontinuation of amiodarone. Dermatologic Effects. Photosensitivity reactions are common. Advise patients to avoid sunlamps and to wear sunscreen and protective clothing when outdoors. With prolonged sun exposure, skin may develop a bluish-gray discoloration, which typically resolves a few months after amiodarone is stopped. Minimizing Adverse Interactions Amiodarone is subject to significant interactions with many drugs. Interactions of special concern are presented here. Drugs Whose Levels Can Be Increased by Amiodarone. Amiodarone can increase levels of several drugs, including quinidine, procainamide, phenytoin, digoxin, diltiazem, warfarin, cyclosporine, and three statins: lovastatin, simvastatin, and atorvastatin. Dosages of these agents often require reduction. Drugs That Can Reduce Amiodarone Levels. Amiodarone levels can be reduced by cholestyramine (which decreases amiodarone absorption) and by agents that induce CYP3A4 (e.g., St. John’s wort, rifampin). Monitor to ensure that amiodarone is still effective. Drugs That Can Increase the Risk of Dysrhythmias. The risk of severe dysrhythmias is increased by diuretics (because they can reduce levels of potassium and magnesium) and by drugs that prolong the QT interval. Drugs That Can Cause Bradycardia. Combining amiodarone with a beta blocker, verapamil, or diltiazem can lead to excessive slowing of heart rate. Grapefruit Juice. Grapefruit juice inhibits CYP3A4 and can raise levels of amiodarone. Toxicity can result. Advise patients to avoid grapefruit juice. Patient education information is highlighted as blue text. a 567 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Lipoproteins are structures that transport lipids (cholesterol and triglycerides [TGs]) in blood. Lipoproteins consist of a hydrophobic core, a hydrophilic shell, plus at least one apolipoprotein, which serves as a recognition site for receptors on cells. Lipoproteins that contain apolipoprotein B-100 transport cholesterol and/or TGs from the liver to peripheral tissues. Lipoproteins that contain apolipoproteins A-I or A-II transport cholesterol from peripheral tissues back to the liver. There are three major types of lipoproteins: VLDLs (verylow-density lipoproteins), LDLs (low-density lipoproteins), and HDLs (high-density lipoproteins). VLDLs transport TGs to peripheral tissues. The contribution of VLDLs to ASCVD is unclear. LDLs transport cholesterol to peripheral tissues. Elevation of LDL cholesterol greatly increases the risk of ASCVD. By reducing LDL cholesterol levels, we can arrest or reverse atherosclerosis, and can thereby reduce morbidity and mortality from ASCVD. HDLs transport cholesterol back to the liver. HDLs protect against ASCVD. Atherogenesis is a chronic inflammatory process that begins with accumulation of LDLs beneath the arterial endothelium, followed by oxidation of LDLs. All adults older than 20 years should be screened every 5 years for total cholesterol, LDL cholesterol, HDL cholesterol, and TGs. Treatment of high LDL cholesterol is based on the individual’s 10-year risk of having a major coronary event. Individuals with established ASCVD or an ASCVD risk equivalent (e.g., diabetes) are in the highest 10-year risk group. Diet modification along with exercise is the primary method for reducing LDL cholesterol. Drugs are employed only if diet modification and exercise fail to reduce LDL cholesterol to the target level. Therapy with cholesterol-lowering drugs must continue lifelong. If these drugs are withdrawn, cholesterol levels will return to pretreatment values. Statins (HMG-CoA reductase inhibitors) are the most effective drugs for lowering LDL cholesterol, and they cause few adverse effects. ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Statins can slow progression of ASCVD, decrease the number of adverse cardiac events, and reduce mortality. Statins reduce LDL cholesterol levels by increasing the number of LDL receptors on hepatocytes, thereby enabling hepatocytes to remove more LDLs from the blood. The process by which LDL receptor number is increased begins with inhibition of HMG-CoA reductase, the rate-limiting enzyme in cholesterol synthesis. Four statins—atorvastatin, fluvastatin, lovastatin, and simvastatin—are metabolized by CYP3A4, and hence their levels can be increased by CYP3A4 inhibitors (e.g., cyclosporine, erythromycin, ketoconazole, ritonavir). Statins can cause liver damage. Tests of liver function should be done at baseline and as clinically indicated thereafter. Statins can cause myopathy. Patients who experience unusual muscle pain, soreness, tenderness, and/or weakness should inform their provider. A marker for muscle injury— creatine kinase (CK)—should be measured at baseline, before starting the drug, and whenever signs or symptoms that could be due to myositis or myopathy develop. Statins should not be used during pregnancy. Bile-acid sequestrants (e.g., colesevelam) reduce LDL cholesterol levels by increasing the number of LDL receptors on hepatocytes. The mechanism is complex and begins with preventing reabsorption of bile acids in the intestine. Bile-acid sequestrants are not absorbed from the GI tract, and hence do not cause systemic adverse effects. However, they can cause constipation and other GI effects. (GI effects with one agent—colesevelam—are minimal.) Older bile-acid sequestrants form complexes with other drugs and thereby prevent their absorption. Accordingly, oral medications should be administered 1 hour before the sequestrant or 4 hours after. With a newer sequestrant— colesevelam—these interactions are minimal. Ezetimibe lowers LDL cholesterol by reducing cholesterol absorption in the small intestine. Like the statins, ezetimibe can cause muscle injury. Gemfibrozil and other fibrates are the most effective drugs for lowering TG levels. Like the statins, the fibrates can cause muscle injury. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa IMPLICATIONS THAT APPLY TO ALL DRUGS THAT LOWER LDL CHOLESTEROL Preadministration Assessment Baseline Data Obtain laboratory values for total cholesterol, LDL cholesterol, HDL cholesterol, and TGs (VLDLs). Identifying ASCVD Risk Factors The patient history and physical examination should identify ASCVD risk factors. These include smoking, advancing age, a personal history of ASCVD, reduced levels of HDL cholesterol (below 40 mg/dL), and hypertension. In the past, diabetes was considered an ASCVD risk factor. However, because the association between diabetes and 588 CHAPTER 50 Prophylaxis of Atherosclerotic Cardiovascular Disease Summary of Major Nursing Implicationsa—cont’d ASCVD is so strong, diabetes is now considered an ASCVD risk equivalent (i.e., it poses the same 10-year risk of a major coronary event as ASCVD itself). Measures to Enhance Therapeutic Effects Diet Modification Diet modification should precede and accompany drug therapy for elevated LDL cholesterol. Inform patients about the importance of diet in controlling cholesterol levels, and arrange for dietary counseling. Advise patients to limit consumption of cholesterol (to below 200 mg/day) and saturated fat (to below 7% of caloric intake). If these measures fail to reduce LDL cholesterol to the target level, advise patients to add soluble fiber and plant stanols or sterols to the regimen. Exercise Regular exercise can reduce LDL cholesterol and elevate HDL cholesterol, reducing the risk of ASCVD. Help the patient establish an appropriate exercise program. Reduction of ASCVD Risk Factors Correctable ASCVD risk factors should be addressed. Encourage smokers to quit. Disease states that promote ASCVD— diabetes mellitus and hypertension—must be treated. Promoting Compliance Drug therapy for elevated LDL cholesterol must continue lifelong; if drugs are withdrawn, cholesterol levels will return to pretreatment values. Inform patients about the need for continuous therapy, and encourage them to adhere to the prescribed regimen. HMG-COA REDUCTASE INHIBITORS (STATINS) Atorvastatin Fluvastatin Lovastatin Pitavastatin Pravastatin Rosuvastatin Simvastatin Exercise caution in patients with nonalcoholic fatty liver disease, in those who consume alcohol to excess, and in those taking fibrates or ezetimibe or agents that inhibit CYP3A4 (e.g., cyclosporine, erythromycin, ketoconazole, ritonavir). Use rosuvastatin with caution in Asian patients. Implementation: Administration Route Oral. Administration Instruct patients to take lovastatin with the evening meal; all other statins can be administered without regard to meals. Advise patients that dosing in the evening is preferred for all statins. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Cholesterol levels should be monitored monthly early in treatment and at longer intervals thereafter. Minimizing Adverse Effects Statins are very well tolerated. Side effects are uncommon, and serious adverse effects—hepatotoxicity and myopathy— are relatively rare. Hepatotoxicity. Statins can injure the liver, but jaundice and other clinical signs are rare. Liver function should be assessed before treatment and as clinically indicated thereafter. If serum transaminase becomes persistently excessive (more than 3 times the ULN), statins should be discontinued. Statins should be avoided in patients with alcoholic or viral hepatitis, but may be used in patients with nonalcoholic fatty liver disease. Myopathy. Statins can cause muscle injury. If statins are not withdrawn, injury may progress to severe myositis or potentially fatal rhabdomyolysis. Inform patients about the risk of myopathy, and instruct them to notify the prescriber if unexplained muscle pain or tenderness develops. If muscle In addition to the implications discussed below, see earlier in this summary for implications that apply to all drugs that lower LDL cholesterol. Preadministration Assessment Therapeutic Goal Statins, in combination with diet modification and exercise, are used primarily to lower levels of LDL cholesterol. Additional indications are shown in Table 50.7. Baseline Data Obtain a baseline lipid profile, consisting of total cholesterol, LDL cholesterol, HDL cholesterol, and TGs (VLDLs). Also, obtain baseline LFTs and a CK level. Identifying High-Risk Patients Statins are contraindicated for patients with viral or alcoholic hepatitis and for women who are pregnant. pain does develop, the CK level should be measured, and if it is more than 10 times the ULN, the statin should be withdrawn or changed. Minimizing Adverse Interactions The risk of myopathy is increased by (1) gemfibrozil, fenofibrate, and ezetimibe, which promote myopathy themselves; and by (2) inhibitors of CYP3A4—such as cyclosporine, macrolide antibiotics (e.g., erythromycin), azole antifungal drugs (e.g., ketoconazole), and HIV protease inhibitors (e.g., ritonavir)—which can cause statin levels to rise. The combination of a statin with any of these drugs should be used with caution. Use in Pregnancy Statins are contraindicated during pregnancy. Inform women of childbearing age about the potential for fetal harm and warn them against becoming pregnant. If pregnancy occurs and the patient intends to continue the pregnancy, statins should be withdrawn. 589 Continued UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d BILE-ACID SEQUESTRANTS GEMFIBROZIL Cholestyramine Colesevelam Colestipol Preadministration Assessment Therapeutic Goal Gemfibrozil, in conjunction with diet modification, is used to reduce elevated levels of TGs (VLDLs). The drug is not very effective at lowering LDL cholesterol. It may also be used to raise low levels of HDL cholesterol. In addition to the implications discussed here, see earlier in this summary for implications that apply to all drugs that lower LDL cholesterol. Preadministration Assessment Therapeutic Goal Bile-acid sequestrants, in conjunction with diet modification and exercise (and a statin if necessary), are used to reduce elevated levels of LDL cholesterol. Baseline Data Obtain laboratory values for total cholesterol, LDL cholesterol, HDL cholesterol, and TGs (VLDLs). Implementation: Administration Route Oral. Baseline Data Obtain laboratory values for total cholesterol, LDL cholesterol, HDL cholesterol, and TGs (VLDLs). Identifying High-Risk Patients Gemfibrozil is contraindicated for patients with liver disease, severe renal dysfunction, and gallbladder disease. Use with caution in patients taking statins or warfarin. Implementation: Administration Route Oral. Administration Administration Instruct patients to mix cholestyramine powder and colestipol granules with water, fruit juice, soup, or pulpy fruit (e.g., applesauce, crushed pineapple) to reduce the risk of esophageal irritation and impaction. Inform patients that the sequestrants are not water soluble, so the mixtures will be cloudy suspensions, not clear solutions. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Cholesterol levels should be monitored monthly early in treatment and at longer intervals thereafter. Minimizing Adverse Effects Constipation. Cholestyramine and colestipol—but not colesevelam—can cause constipation. Inform patients that constipation can be minimized by increasing dietary fiber and fluids. A mild laxative may be used if needed. Instruct patients taking cholestyramine or colestipol to notify the prescriber if constipation becomes bothersome, in which case a switch to colesevelam should be considered. Vitamin Deficiency. Cholestyramine and colestipol—but not colesevelam—can impair absorption of fat-soluble vitamins (A, D, E, and K). Vitamin supplements may be required. Colesevelam does not reduce vitamin absorption. Minimizing Adverse Interactions Cholestyramine and colestipol—but not colesevelam—can bind with other drugs and prevent their absorption. Advise patients to administer other medications 1 hour before these sequestrants or 4 hours after. Instruct patients to administer gemfibrozil 30 minutes before the morning and evening meals. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Obtain periodic tests of blood lipids. Minimizing Adverse Effects Gallstones. Gemfibrozil increases gallstone development. Inform patients about symptoms of gallbladder disease (e.g., upper abdominal discomfort, intolerance of fried foods, bloating), and instruct them to notify the prescriber if these develop. Myopathy. Gemfibrozil can cause muscle damage. Warn patients to report any signs of muscle injury, such as tenderness, weakness, or unusual muscle pain. Liver Disease. Gemfibrozil may disrupt liver function. Cancer of the liver may also be a risk. Obtain periodic tests of liver function. Minimizing Adverse Interactions Warfarin. Gemfibrozil enhances the effects of warfarin, thereby increasing the risk of bleeding. Obtain more frequent measurements of prothrombin time and assess the patient for signs of bleeding. Reduction of warfarin dosage may be required, and reassessment and readjustment of the warfarin dosage may be needed if the fibrate is stopped. Statins. Gemfibrozil and statins both cause muscle injury. Risks rise when both are used. Use the combination with caution. Patient education information is highlighted as blue text. a 590 CHAPTER 51 reduce cholesterol reduce that risk. Accordingly, all patients with high cholesterol levels should receive cholesterol-lowering therapy. Hypertension. High blood pressure increases the risk of cardiovascular mortality, and lowering blood pressure reduces the risk. Accordingly, all patients with hypertension should receive treatment. Blood pressure should be reduced to 140/90 mm Hg or less. In patients with additional risk factors (e.g., diabetes, heart failure, retinopathy), the target blood pressure is 130/80 mm Hg or less. Management of hypertension is discussed in Chapter 47. Diabetes. Both type 1 (insulin-dependent) and type 2 (non–insulin-dependent) diabetes increase the risk of cardiovascular mortality. Type 1 increases the risk 3- to 10-fold; type 2 increases the risk 2- to 4-fold. Although there is good evidence that tight glycemic control decreases the risk of microvascular complications of diabetes, there is little evidence to show that tight glycemic control decreases the risk of cardiovascular complications. Nonetheless, it is prudent to strive for optimal glycemic control. Drugs for Angina Pectoris Physical Inactivity. Increased physical activity has multiple benefits. In patients with chronic stable angina, exercise increases exercise tolerance and the sense of well-being, and decreases anginal symptoms, cholesterol levels, and objective measures of ischemia. Accordingly, the guidelines recommend that patients perform 30 to 60 minutes of a moderate-intensity activity 3 to 4 times a week. Such activities include walking, jogging, cycling, and other aerobic exercises. Exercise by moderate- to high-risk patients should be medically supervised. Management of Variant Angina Treatment of vasospastic angina can proceed in three steps. For initial therapy, either a calcium channel blocker or a longacting nitrate is selected. If either drug alone is inadequate, then combined therapy with a calcium channel blocker plus a nitrate should be tried. If the combination fails to control symptoms, CABG surgery may be indicated. Beta blockers are not effective in vasospastic angina. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Anginal pain occurs when cardiac oxygen supply is insufficient to meet cardiac oxygen demand. Cardiac oxygen demand is determined by heart rate, contractility, preload, and afterload. Drugs that reduce these factors can help relieve anginal pain. Cardiac oxygen supply is determined by myocardial blood flow. Drugs that increase oxygen supply will reduce anginal pain. Angina pectoris has three forms: chronic stable angina, variant (vasospastic) angina, and unstable angina. The underlying cause of stable angina is coronary artery atherosclerosis. The underlying cause of variant angina is coronary artery spasm. Drugs relieve pain of stable angina by decreasing cardiac oxygen demand. They do not increase oxygen supply. Drugs relieve pain of variant angina by increasing cardiac oxygen supply. They do not decrease oxygen demand. Nitroglycerin and other organic nitrates are vasodilators. To cause vasodilation, nitroglycerin must first be converted to nitric oxide, its active form. This reaction requires a sulfhydryl source. Nitroglycerin relieves pain of stable angina by dilating veins, which decreases venous return, which decreases preload, which decreases oxygen demand. Nitroglycerin relieves pain of variant angina by relaxing coronary vasospasm, which increases oxygen supply. Nitroglycerin is highly lipid soluble, and therefore is readily absorbed through the skin and oral mucosa. Nitroglycerin undergoes very rapid inactivation in the liver. Hence, when the drug is administered orally, most of each dose is destroyed before reaching the systemic circulation. When nitroglycerin is administered sublingually, it is absorbed directly into the systemic circulation and therefore ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ temporarily bypasses the liver. Hence, to produce equivalent effects, sublingual doses can be much smaller than oral doses. Nitroglycerin causes three characteristic side effects: headache, orthostatic hypotension, and reflex tachycardia. All three occur secondary to vasodilation. Reflex tachycardia from nitroglycerin can be prevented with a beta blocker, verapamil, or diltiazem. Continuous use of nitroglycerin can produce tolerance within 24 hours. The mechanism may be depletion of sulfhydryl groups. To prevent tolerance, nitroglycerin should be used in the lowest effective dosage, and long-acting formulations should be used on an intermittent schedule that allows at least 8 drug-free hours every day, usually during the night. Nitroglycerin preparations that have a rapid onset (e.g., sublingual nitroglycerin) are used to abort an ongoing anginal attack and to provide acute prophylaxis when exertion is expected. Administration is PRN. Nitroglycerin preparations that have a long duration (e.g., patches, sustained-release oral capsules) are used for extended protection against anginal attacks. Administration is on a fixed schedule (but one that allows at least 8 drugfree hours a day). Nitroglycerin should be used cautiously with most vasodilators and must not be used at all with sildenafil [Viagra] and other PDE5 inhibitors. Beta blockers prevent pain of stable angina primarily by decreasing heart rate and contractility, which reduces cardiac oxygen demand. Beta blockers are administered on a fixed schedule, not PRN. Beta blockers are not used for variant angina. CCBs relieve the pain of stable angina by reducing cardiac oxygen demand. Two mechanisms are involved. First, all Continued 601 UNIT VII ■ ■ ■ ■ ■ Drugs That Affect the Heart, Blood Vessels, and Blood CCBs relax peripheral arterioles and decrease afterload. Second, verapamil and diltiazem reduce heart rate and contractility (in addition to decreasing afterload). CCBs relieve pain of variant angina by increasing cardiac oxygen supply. The mechanism is relaxation of coronary artery spasm. When a CCB is combined with a beta blocker, a dihydropyridine (e.g., nifedipine) is preferred to verapamil or diltiazem. Verapamil and diltiazem will intensify cardiosuppression caused by the beta blocker, whereas a dihydropyridine will not. Ranolazine appears to reduce anginal pain by helping the heart generate energy more efficiently. Ranolazine should not be used alone. Rather, it should be combined with a nitrate, a beta blocker, or the CCB amlodipine. Ranolazine increases the QT interval and may pose a risk of torsades de pointes, a serious ventricular dysrhythmia. ■ ■ ■ ■ In patients with chronic stable angina, treatment has two objectives: (1) prevention of MI and death and (2) prevention of anginal pain. The risk of MI and death can be decreased with two types of drugs: (1) antiplatelet agents (e.g., aspirin, clopidogrel) and (2) cholesterol-lowering drugs. Anginal pain is prevented with one or more long-acting antianginal drugs (beta blocker, CCB, long-acting nitrate) supplemented with sublingual nitroglycerin when breakthrough pain occurs. As a rule, revascularization with CABG surgery or PCI is indicated only after treatment with two or three antianginal drugs has failed. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa NITROGLYCERIN Preadministration Assessment Therapeutic Goal Reduction of the frequency and intensity of anginal attacks. Baseline Data Obtain baseline data on the frequency and intensity of anginal attacks, the location of anginal pain, and the factors that precipitate attacks. The patient interview and physical examination should identify risk factors for angina pectoris, including treatable contributing pathophysiologic conditions (e.g., hypertension, hyperlipidemia). Identifying High-Risk Patients Use with caution in hypotensive patients and in patients taking drugs that can lower blood pressure, including alcohol and antihypertensive medications. Use with sildenafil [Viagra] and other PDE5 inhibitors is contraindicated. Implementation: Administration Routes and Administration Sublingual Tablets or Powder Use. Prophylaxis or termination of an acute anginal attack. Technique of Administration. Instruct patients to place the tablet or empty the powder under the tongue and leave it there until fully dissolved; these medications should not be swallowed. Instruct patients to call 911 or go to an emergency department if pain is not relieved in 5 minutes. While awaiting emergency care, they can take 1 more dose, and then a third 5 minutes later. Instruct patients to store tablets in a dry place at room temperature in their original container, which should be closed tightly after each use. Under these conditions, the tablets should remain effective until the expiration date on the container. Sustained-Release Oral Capsules Use. Sustained protection against anginal attacks. To avoid tolerance, administer only once or twice daily. Technique of Administration. Instruct patients to swallow these preparations intact, without chewing or crushing. Transdermal Delivery Systems Use. Sustained protection against anginal attacks. Technique of Administration. Instruct patients to apply transdermal patches to a hairless area of skin, using a new patch and a different site each day. Instruct patients to remove the patch after 12 to 14 hours, allowing 10 to 12 “patch-free” hours each day. This will prevent tolerance. Translingual Spray Use. Prophylaxis or termination of an acute anginal attack. Technique of Administration. Instruct patients to direct the spray against the oral mucosa. Warn patients not to inhale the spray. Topical Ointment Use. Sustained protection against anginal attacks. Instruct patients to remove any remaining ointment before applying a new dose. Technique of Administration. (1) Squeeze a ribbon of ointment of prescribed length onto the applicator paper provided; (2) using the applicator paper, spread the ointment over an area at least 2.5 inches by 3.5 inches (application may be made to the chest, back, abdomen, upper arm, or anterior thigh); and (3) cover the ointment with plastic wrap. Avoid touching the ointment. Instruct patients to rotate the application site to minimize local irritation. 602 CHAPTER 51 Drugs for Angina Pectoris Summary of Major Nursing Implicationsa—cont’d Intravenous Uses. (1) Angina pectoris refractory to more conventional therapy, (2) perioperative control of blood pressure, (3) production of controlled hypotension during surgery, and (4) heart failure associated with acute MI. Technique of Administration. Infuse IV using a glass IV bottle and the administration set provided by the manufacturer; avoid standard IV tubing. Check the stock solution label to verify volume and concentration, which can differ among manufacturers. Dilute stock solutions before use. Administer by continuous infusion. The rate is slow initially (5 mcg/min) and then gradually increased until an adequate response is achieved. Monitor cardiovascular status constantly. Minimizing Adverse Effects Headache. Inform patients that headache will diminish with continued drug use. Advise patients that headache can be relieved with aspirin, acetaminophen, or some other mild analgesic. Orthostatic Hypotension. Inform patients about symptoms of hypotension (e.g., dizziness, light-headedness), and advise them to sit or lie down if these occur. Inform patients that hypotension can be minimized by moving slowly when changing from a sitting or supine position to an upright posture. Reflex Tachycardia. This reaction can be suppressed by concurrent treatment with a beta blocker, verapamil, or diltiazem. Terminating Therapy Minimizing Adverse Interactions Warn patients against abrupt withdrawal of long-acting preparations (transdermal systems, topical ointment, sustainedrelease tablets and capsules). Hypotensive Agents, Including PDE5 Inhibitors. Nitroglycerin can interact with other hypotensive drugs to produce excessive lowering of blood pressure. Advise patients to avoid alcohol. Exercise caution when nitroglycerin is used in combination with beta blockers, calcium channel blockers, diuretics, and all other drugs that can lower blood pressure. Implementation: Measures to Enhance Therapeutic Effects Reducing Risk Factors Precipitating Factors. Advise patients to avoid activities that are likely to elicit an anginal attack (e.g., overexertion, heavy meals, emotional stress, cold exposure). Exercise. Encourage patients who have a sedentary lifestyle to establish a regular program of aerobic exercise (e.g., walking, jogging, swimming, biking). Smoking Cessation. Strongly encourage patients to quit smoking. Contributing Disease States. Ensure that patients with contributing pathology (especially hypertension or hypercholesterolemia) are receiving appropriate treatment. Warn patients not to combine nitroglycerin with a PDE5 inhibitor (e.g., sildenafil [Viagra]), because life-threatening hypotension can result. ISOSORBIDE MONONITRATE AND ISOSORBIDE DINITRATE Both drugs have pharmacologic actions identical to those of nitroglycerin. Differences relate only to dosage forms, routes of administration, and time course of action. Therefore, the implications presented for nitroglycerin apply to these drugs as well. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Instruct patients to keep a record of the frequency and intensity of anginal attacks, the location of anginal pain, and the factors that precipitate attacks. Patient education information is highlighted as blue text. a 603 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood with respect to mortality (6.2% with each drug), ICH (0.93% with tenecteplase vs. 0.94% with tPA), and total stroke (1.78% vs. 1.66% with tPA). Of significance, the incidence of major hemorrhage (other than intracranial) was lower with tenecteplase (4.7% vs. 5.9%). Tenecteplase dosage is based on body weight as follows: • • • • • Below 60 kg: dose 30 mg 60 to 69.9 kg: dose 35 mg 70 to 79.9 kg: dose 40 mg 80 to 89.9 kg: dose 45 mg Above 90 kg: dose 50 mg Reteplase Reteplase [Retavase] is a derivative of tPA produced by recombinant DNA technology. In contrast to tPA itself, which contains 527 amino acids, reteplase is composed of only 355 amino acids. Like tPA, reteplase converts plasminogen to plasmin, which in turn digests the fibrin matrix of the thrombus. Reteplase has a short half-life (13 to 16 minutes), owing to rapid clearance by the liver and kidneys. As with other thrombolytic drugs, bleeding is the major adverse effect. The risk of bleeding is increased by concurrent use of heparin, aspirin, and other drugs that impair hemostasis. Reteplase is approved only for acute MI. Treatment consists of two 10-unit doses separated by 30 minutes. Each dose is given by IV bolus injected over a 2-minute interval. Reteplase should not be administered through a line that contains heparin. If a heparin-containing line must be used, it should be flushed before giving reteplase. KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Hemostasis occurs in two stages: formation of a platelet plug, followed by coagulation (i.e., production of fibrin, a protein that reinforces the platelet plug). Platelet aggregation depends on activation of platelet glycoprotein (GP) IIb/IIIa receptors, which bind fibrinogen to form cross-links between platelets. Fibrin is produced by two pathways—the contact activation pathway (aka intrinsic pathway) and the tissue factor pathway (aka extrinsic pathway)—that converge at clotting factor Xa, which catalyzes formation of thrombin, which in turn catalyzes formation of fibrin. Four factors in the coagulation pathways require an activated form of vitamin K for their synthesis. Plasmin, the active form of plasminogen, serves to degrade the fibrin meshwork of clots. A thrombus is a blood clot formed within a blood vessel or the atria of the heart. Arterial thrombi begin with formation of a platelet plug, which is then reinforced with fibrin. Venous thrombi begin with formation of fibrin, which then enmeshes red blood cells and platelets. Arterial thrombi are best prevented with antiplatelet drugs (e.g., aspirin, clopidogrel), whereas venous thrombi are best prevented with anticoagulants (e.g., heparin, warfarin, dabigatran). Heparin is a large polymer (molecular weight range, 3000 to 30,000) that carries many negative charges. Heparin suppresses coagulation by helping antithrombin inactivate thrombin and factor Xa. Heparin is administered IV or subQ. Because of its large size and negative charges, heparin is unable to cross membranes, and hence cannot be administered PO. Anticoagulant effects of heparin develop within minutes of IV administration. The major adverse effect of heparin is bleeding. Severe heparin-induced bleeding can be treated with protamine sulfate, a drug that binds heparin and thereby stops it from working. Heparin-induced thrombocytopenia is a potentially fatal condition caused by development of antibodies against heparin–platelet protein complexes. Heparin is contraindicated for patients with thrombocytopenia or uncontrollable bleeding, and must be used with ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ 628 extreme caution in all patients for whom there is a high likelihood of bleeding. Heparin therapy is monitored by measuring the activated partial thromboplastin time (aPTT) or anti-Xa heparin assay. The target aPTT is 60 to 80 seconds (i.e., 1.5 to 2 times the normal value of 40 seconds). The target anti-Xa level is 0.3 to 0.7 IU/mL. Low-molecular-weight (LMW) heparins are produced by breaking molecules of unfractionated heparin into smaller pieces. In contrast to unfractionated heparin, which inactivates factor Xa and thrombin equally, LMW heparins preferentially inactivate factor Xa. In contrast to unfractionated heparin, LMW heparins do not bind nonspecifically to plasma proteins and tissues. As a result, their bioavailability is high, making their plasma levels predictable. Because plasma levels of LMW heparins are predictable, these drugs can be administered using a fixed dosage, with no need for routine laboratory monitoring. As a result, LMW heparins can be used at home. Warfarin is our oldest oral anticoagulant. Warfarin prevents the activation of vitamin K and thereby blocks the biosynthesis of vitamin K–dependent clotting factors. Anticoagulant responses to warfarin develop slowly and persist for several days after warfarin is discontinued. Warfarin is used to prevent venous thromboembolism (VTE) and to prevent stroke and systemic embolism in patients with atrial fibrillation. Warfarin therapy is monitored by measuring prothrombin time (PT). Results are expressed as an international normalized ratio (INR). An INR of 2 to 3 is the target for most patients. Bleeding is the major complication of warfarin therapy. Genetic testing for variant genes that code for VKORC1 and CYP2C9 can identify people with increased sensitivity to warfarin, and who therefore may need a dosage reduction. Moderate warfarin overdose is treated with vitamin K. Warfarin must not be used during pregnancy. The drug can cause fetal malformation, CNS defects, and optic atrophy. Warfarin is subject to a large number of clinically significant drug interactions. Drugs can increase anticoagulant effects CHAPTER 52 ■ ■ ■ ■ ■ ■ ■ ■ ■ by displacing warfarin from plasma albumin, by inhibiting hepatic enzymes that degrade warfarin, and by decreasing synthesis of clotting factors. Drugs can decrease anticoagulant effects by inducing hepatic drug-metabolizing enzymes, increasing synthesis of clotting factors, and inhibiting warfarin absorption. Drugs that promote bleeding, such as heparin and aspirin, will obviously increase the risk of bleeding in patients taking warfarin. Instruct patients to avoid all drugs—prescription and nonprescription—that have not been specifically approved by the prescriber. Dabigatran is an oral anticoagulant that works by direct inhibition of thrombin. Dabigatran is an alternative to warfarin for chronic anticoagulation in patients with atrial fibrillation. Compared with warfarin, dabigatran has five advantages: rapid onset, fixed dosage, no need for coagulation testing, few drug-food interactions, and a lower risk of hemorrhagic stroke and other major bleeds. Compared with warfarin, dabigatran has three disadvantages: no antidote, limited clinical experience, and more GI disturbances (dyspepsia, ulceration, gastritis, etc.). Rivaroxaban, edoxaban, and apixaban are oral anticoagulants that work by direct inhibition of factor Xa. Like dabigatran, rivaroxaban, edoxaban, and apixaban are safer than warfarin and easier to use. Aspirin and other antiplatelet drugs suppress thrombus formation in arteries. Aspirin inhibits platelet aggregation by causing irreversible inhibition of cyclooxygenase. Since platelets are unable to synthesize new cyclooxygenase, inhibition persists for the life of the platelet (7 to 10 days). In its role as an antiplatelet drug, aspirin is given for multiple purposes, including primary prevention of myocardial ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Anticoagulant, Antiplatelet, and Thrombolytic Drugs infarction (MI), acute management of MI, and reduction of cardiovascular events in patients with unstable angina, chronic stable angina, ischemic stroke, or a history of transient ischemic attacks (TIAs). When used to suppress platelet aggregation, aspirin is administered in low doses—typically 80 to 325 mg/day. Clopidogrel suppresses platelet aggregation by causing irreversible blockade of P2Y12 ADP receptors on the platelet surface. Clopidogrel is a prodrug that undergoes conversion to its active form by hepatic CYP2C19. Patients with an inherited deficiency in CYP2C19 may have an unreliable response to clopidogrel. The major adverse effect of clopidogrel is bleeding. The GP IIb/IIIa receptor blockers (e.g., abciximab) inhibit the final common step in platelet aggregation, and hence are the most effective antiplatelet drugs available. Alteplase (tPA) and other thrombolytic drugs (aka fibrinolytic drugs) are used to dissolve existing thrombi (rather than prevent thrombi from forming). Thrombolytic drugs work by converting plasminogen to plasmin, an enzyme that degrades the fibrin matrix of thrombi. Thrombolytic therapy is most effective when started early (e.g., for acute MI, within 4 to 6 hours of symptom onset, and preferably sooner). Thrombolytic drugs carry a significant risk of bleeding. Intracranial hemorrhage is the greatest concern. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa HEPARIN Preadministration Assessment Therapeutic Goal The objective is to prevent thrombosis without inducing spontaneous bleeding. Heparin is the preferred anticoagulant for use during pregnancy and in situations that require rapid onset of effects, including PE, evolving stroke, and massive DVT. Other indications include open heart surgery, renal dialysis, and disseminated intravascular coagulation. Low doses are used to prevent postoperative venous thrombosis and to enhance thrombolytic therapy of MI. Baseline Data Obtain baseline values for blood pressure, heart rate, complete blood cell counts, platelet counts, hematocrit, and aPTT. Identifying High-Risk Patients Heparin is contraindicated for patients with severe thrombocytopenia or uncontrollable bleeding and for patients undergoing lumbar puncture, regional anesthesia, or surgery of the eye, brain, or spinal cord. Use with extreme caution in patients at high risk of bleeding, including those with hemophilia, increased capillary permeability, dissecting aneurysm, GI ulcers, or severe hypertension. Caution is also needed in patients with severe hepatic or renal impairment. Implementation: Administration Routes Intravenous (continuous infusion or intermittent) and subQ. Avoid IM injections! Administration General Considerations. Dosage is prescribed in units, not milligrams. Heparin preparations vary widely in concentration; read the label carefully to ensure correct dosing. Continuous IV Infusion. Administer with a continuous infusion pump or some other approved volume-control unit. Policy may require that dosage be double-checked by a second person. Check the infusion rate every 30 to 60 minutes. During Continued 629 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d the early phase of treatment, the aPTT or anti-Xa level should be determined every 6 hours. Check the site of needle insertion periodically for extravasation. Ongoing Evaluation and Interventions Evaluating Treatment We evaluate treatment by measuring the aPTT or the anti-Xa level. Heparin should increase the aPTT by 1.5- to 2-fold above baseline. Therapeutic range for anti-Xa level is 0.3 to 0.7 IU/mL. Minimizing Adverse Effects Hemorrhage. Heparin overdose may cause hemorrhage. Monitor closely for signs of bleeding. These include reduced blood pressure, elevated heart rate, discolored urine or stool, bruises, petechiae, hematomas, persistent headache or faintness (suggestive of cerebral hemorrhage), pelvic pain (suggestive of ovarian hemorrhage), and lumbar pain (suggestive of adrenal hemorrhage). Laboratory data suggesting hemorrhage include reductions in the hematocrit and blood cell counts. If bleeding occurs, heparin should be discontinued. Severe overdose can be treated with protamine sulfate administered by slow IV injection. The risk of bleeding can be reduced by ensuring that the aPTT or the anti-Xa levels are not above recommended range according to facility protocol. Heparin-Induced Thrombocytopenia. HIT, characterized by reduced platelet counts and increased thrombotic events, poses a risk of DVT, PE, cerebral thrombosis, MI, and ischemic injury to the arms and legs. To reduce risk, monitor platelet counts 2 to 3 times a week during the first 3 weeks of heparin use, and monthly thereafter. If severe thrombocytopenia develops (platelet count below 100,000/ mm3), discontinue heparin and, if anticoagulation is still needed, substitute another anticoagulant, such as argatroban. Spinal/Epidural Hematoma. Heparin and all other anticoagulants pose a risk of spinal or epidural hematoma in patients undergoing spinal puncture or spinal/epidural anesthesia. Prolonged or permanent paralysis can result. Risk of hematoma is increased by several factors, including use of an indwelling epidural catheter, use of other anticoagulants (e.g., warfarin, dabigatran), and use of antiplatelet drugs (e.g., aspirin, clopidogrel). Monitor for signs and symptoms of neurologic impairment. If impairment develops, immediate intervention is needed. Hypersensitivity Reactions. Allergy may develop to antigens in heparin preparations. To minimize the risk of severe reactions, administer a small test dose before the full therapeutic dose. Minimizing Adverse Interactions Antiplatelet Drugs. Concurrent use of aspirin, clopidogrel, and other antiplatelet drugs increases the risk of bleeding. Use these agents with caution. WARFARIN, A VITAMIN K ANTAGONIST Preadministration Assessment Therapeutic Goal The goal is to prevent thrombosis without inducing spontaneous bleeding. Specific indications include prevention of venous thrombosis and associated PE, prevention of thromboembolism in patients with prosthetic heart valves, and prevention of stroke and systemic embolism in patients with atrial fibrillation. Baseline Data Obtain a thorough medical history. Be sure to identify use of any medications that might interact adversely with warfarin. Obtain baseline values of vital signs and PT. Genetic testing for variants of CYP2C9 and VKORC1 may be done to identify patients who may require a reduction in warfarin dosage. Identifying High-Risk Patients Warfarin is contraindicated in the presence of vitamin K deficiency, liver disease, alcoholism, thrombocytopenia, uncontrollable bleeding, pregnancy, and lactation, and for patients undergoing lumbar puncture, regional anesthesia, or surgery of the eye, brain, or spinal cord. Use with extreme caution in patients at high risk of bleeding, including those with hemophilia, increased capillary permeability, dissecting aneurysm, GI ulcers, and severe hypertension. Use with caution in patients with variant forms of CYP2C9 or VKORC1. Implementation: Administration Route Oral. Administration For most patients, dosage is adjusted to maintain an INR value of 2 to 3. Maintain a flow chart for hospitalized patients indicating INR values, dose, and administration time. Implementation: Measures to Enhance Therapeutic Effects Promoting Adherence Safe and effective therapy requires rigid adherence to the dosing schedule. Achieving adherence requires active and informed participation by the patient. Provide the patient with detailed written and verbal instructions regarding the purpose of treatment, dosage size and timing, and the importance of careful adherence to the dosing schedule. Also, provide the patient with a chart on which to keep an ongoing record of warfarin use. If the patient is incompetent (e.g., mentally ill, alcoholic, senile), ensure that a responsible individual supervises treatment. 630 CHAPTER 52 Anticoagulant, Antiplatelet, and Thrombolytic Drugs Summary of Major Nursing Implicationsa—cont’d Nondrug Measures Advise the patient to (1) avoid prolonged immobility, (2) elevate the legs when sitting, (3) avoid garments that can restrict blood flow in the legs, (4) participate in exercise activities, and (5) wear support hose. These measures will reduce venous stasis and will thereby reduce the risk of thrombosis. Ongoing Evaluation and Interventions Monitoring Treatment Evaluate therapy by monitoring PT. Test results are reported as an international normalized ratio (INR). For most patients, the target INR is 2 to 3. If the INR is below this range, dosage should be increased. Conversely, if the INR is above this range, dosage should be reduced. The INR should be determined frequently: daily during the first 5 days, twice a week for the next 1 to 2 weeks, once a week for the next 1 to 2 months, and every 2 to 4 weeks thereafter. In addition, the INR should be determined whenever a drug that interacts with warfarin is added to or withdrawn from the regimen. When appropriate, teach patients how to monitor their PT and INR at home. Minimizing Adverse Effects Hemorrhage. Hemorrhage is the major complication of warfarin therapy. Warn patients about the danger of hemorrhage, and inform them about signs of bleeding. These include reduced blood pressure, elevated heart rate, discolored urine or stools, bruises, petechiae, hematomas, persistent headache or faintness (suggestive of cerebral hemorrhage), pelvic pain (suggestive of ovarian hemorrhage), and lumbar pain (suggestive of adrenal hemorrhage). Laboratory data suggesting hemorrhage include reductions in the hematocrit and blood cell counts. Instruct the patient to withhold warfarin and notify the prescriber if signs of bleeding are noted. Advise the patient to wear some form of identification (e.g., Medic Alert bracelet) to alert emergency personnel to warfarin use. To reduce the incidence of bleeding, advise the patient to avoid excessive consumption of alcohol. Suggest use of a soft toothbrush to prevent bleeding from the gums. Advise patients to shave with an electric razor. Warfarin intensifies bleeding during surgical procedures. Instruct the patient to make certain the surgeon is aware of warfarin use. Warfarin should be discontinued several days before elective procedures. If emergency surgery must be performed, vitamin K1 can help reduce bleeding. Warfarin-induced bleeding can be controlled with vitamin K1. For most patients, oral vitamin K will suffice. For patients with severe bleeding or a very high INR, vitamin K is given by injection (usually IV). Use in Pregnancy and Lactation. Warfarin can cross the placenta, causing fetal hemorrhage and malformation. Inform those of childbearing age about potential risks to the fetus, and warn them against becoming pregnant. If pregnancy develops, termination should be considered. Warfarin enters breast milk and may harm the nursing infant. Warn patients against breast-feeding. Minimizing Adverse Interactions Inform patients that warfarin is subject to a large number of potentially dangerous drug interactions. Instruct them to avoid all drugs—prescription and nonprescription—that have not been specifically approved by the prescriber. Before treatment, take a complete medication history to identify any drugs that might interact adversely with warfarin. CLOPIDOGREL, A P2Y12 ADENOSINE DIPHOSPHATE RECEPTOR ANTAGONIST Preadministration Assessment Therapeutic Goal Clopidogrel is used to prevent blockage of coronary artery stents and to reduce thrombotic events—MI, ischemic stroke, and vascular death—in patients with ACS and in those with atherosclerosis documented by recent MI, recent stroke, or established peripheral arterial disease. Baseline Data Consider testing for variants of the CYP2C19 gene to determine whether the patient is a poor metabolizer of clopidogrel. Identifying High-Risk Patients Clopidogrel is contraindicated in patients with active pathologic bleeding, including ICH and bleeding ulcers. Use with caution in patients taking other drugs that promote bleeding. Generally avoid clopidogrel in poor metabolizers of the drug. Implementation: Administration Route Oral. Administration Instruct patients to take clopidogrel once a day, with or without food. Ongoing Evaluation and Interventions Promoting Beneficial Effects Instruct patients being treated for ACS to take aspirin (75 to 325 mg) once daily. Minimizing Adverse Effects Bleeding. Clopidogrel poses a risk of serious bleeding. Avoid clopidogrel in patients with active pathologic bleeding, and use with caution in patients taking other drugs that promote bleeding. If possible, manage major bleeding without stopping clopidogrel, since discontinuation would increase the risk of a thrombotic event. Inform patients about the risk of bleeding, and warn them that: • You may bruise more easily. • You may bleed more easily. Continued 631 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d • You are more likely to get nosebleeds. • Bleeding will take longer than usual to stop. Instruct patients to contact the prescriber if they experience any of these symptoms of bleeding: • Unexpected bleeding • Bleeding that lasts a long time • Blood in the urine, indicated by discoloration (pink, red, brown) • Blood in stools (indicated by red, black, tarry stools) • Bruising with no obvious cause • Vomiting blood, which may look like coffee grounds Instruct patients that, even if these symptoms occur, they should continue taking clopidogrel until the prescriber says they should stop. Instruct patients to discontinue clopidogrel 5 days before elective surgery. Thrombotic Thrombocytopenic Purpura (TTP). Rarely, patients develop TTP, a potentially fatal condition characterized by thrombocytopenia, hemolytic anemia, neurologic symptoms, renal dysfunction, and fever. If TTP is diagnosed, urgent treatment—including plasmapheresis—is required. Minimizing Adverse Interactions Drugs That Promote Bleeding. Use with caution in patients taking other drugs that promote bleeding (e.g., heparin, warfarin, dabigatran, aspirin, and nonaspirin NSAIDs). Proton Pump Inhibitors (PPIs). The PPIs can help prevent clopidogrel-related GI bleeding—but may reduce the benefits of clopidogrel by inhibiting CYP2C19, the hepatic enzyme that converts clopidogrel to its active form. In patients with risk factors for GI bleeding (e.g., advanced age, use of NSAIDs or anticoagulants), the benefits of combining a PPI with clopidogrel probably outweigh any risk from reduced antiplatelet effects. Conversely, in patients who lack risk factors for GI bleeding, combined use of clopidogrel with a PPI may reduce the benefits of clopidogrel without offering any meaningful GI protection—and hence combining a PPI with clopidogrel in these patients should probably be avoided. When a PPI is used with clopidogrel, pantoprazole is a good choice because, compared with other PPIs, pantoprazole causes less inhibition of CYP2C19. CYP2C19 Inhibitors (Other Than PPIs). Like the PPIs, several other drugs can inhibit CYP2C19. Among these are cimetidine, fluoxetine, fluvoxamine, fluconazole, ketoconazole, voriconazole, etravirine, felbamate, and ticlopidine. Since these drugs may reduce the antiplatelet effects of clopidogrel, using an alternative to these drugs is preferred. Preadministration Assessment Therapeutic Goal All three thrombolytic drugs are used for acute MI, and one drug—alteplase—is also used for ischemic stroke and PE, and, in low dosage, for clearing blocked central venous catheters. Baseline Data Obtain baseline values for blood pressure, heart rate, platelet counts, hematocrit, aPTT, PT, and fibrinogen level. Identifying High-Risk Patients Thrombolytic drugs are contraindicated for patients with active bleeding, aortic dissection, acute pericarditis, cerebral neoplasm, cerebrovascular disease, or a history of intracranial bleeding. Use with great caution in patients with relative contraindications, including pregnancy, severe hypertension, ischemic stroke within the prior 6 months, and major surgery within the prior 2 to 4 weeks. See Table 52.10 for other absolute and relative contraindications. Implementation: Administration Route Intravenous. Administration (for Acute MI) Alteplase. Administer as an initial IV bolus followed by a 90-minute IV infusion. Tenecteplase. Administer as a single IV bolus. Reteplase. Administer as two IV boluses, separated by 30 minutes. Ongoing Evaluation and Interventions Minimizing Adverse Effects Hemorrhage. Thrombolytics may cause bleeding; ICH is the greatest concern. To reduce the risk of major bleeding, minimize manipulation of the patient, avoid subQ and IM injections, minimize invasive procedures, and minimize concurrent use of anticoagulants and antiplatelet drugs. Manage oozing at cutaneous puncture sites with a pressure dressing. Minimizing Adverse Interactions Anticoagulants and Antiplatelet Drugs. Anticoagulants (e.g., heparin, warfarin, dabigatran) and antiplatelet drugs (e.g., aspirin, clopidogrel) increase the risk of bleeding from antithrombotics. Avoid high-dose therapy with these drugs until thrombolytic effects have subsided. THROMBOLYTIC (FIBRINOLYTIC) DRUGS Alteplase (tPA) Reteplase Tenecteplase Patient education information is highlighted as blue text. a 632 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Ventricular Dysrhythmias These dysrhythmias develop frequently and are the major cause of death following MI. Sudden death from dysrhythmias occurs in 15% of patients during the first hour. Ultimately, ventricular dysrhythmias cause 60% of infarction-related deaths. Acute management of ventricular fibrillation consists of defibrillation followed by IV amiodarone for 24 to 48 hours. Programmed ventricular stimulation with guided antidysrhythmic therapy may be lifesaving for some patients. Attempts to prevent dysrhythmias by giving antidysrhythmic drugs prophylactically have failed to reduce mortality. Worse yet, attempted prophylaxis of ventricular dysrhythmias with two drugs—encainide and flecainide—actually increased mortality. Similarly, when quinidine was employed to prevent supraventricular dysrhythmias, it too increased mortality. Therefore, since prophylaxis with antidysrhythmic drugs does not reduce mortality—and may in fact increase mortality— antidysrhythmic drugs should be withheld until a dysrhythmia actually occurs. Cardiogenic Shock Shock results from greatly reduced tissue perfusion secondary to impaired cardiac function. Shock develops in 7% to 10% of patients in the first few days after MI and has a mortality rate of up to 50% in hospitalized patients. Patients at highest risk are those with large infarcts, a previous infarct, a low ejection fraction (less than 35%), diabetes, and advanced age. Drug therapy includes inotropic agents (e.g., dopamine, dobutamine) to increase cardiac output and vasodilators (nitroglycerin, nitroprusside) to improve tissue perfusion and to reduce cardiac work and oxygen demand. Unfortunately, although these drugs can improve hemodynamic status, they do not seem to reduce mortality. Restoration of cardiac perfusion with PCI or coronary artery bypass grafting may be of value. Heart Failure Heart failure secondary to acute MI can be treated with a combination of drugs. A diuretic (e.g., furosemide) is given to decrease preload and pulmonary congestion. Inotropic agents (e.g., digoxin) increase cardiac output by enhancing contractility. Vasodilators (e.g., nitroglycerin, nitroprusside) improve hemodynamic status by reducing preload, afterload, or both. ACE inhibitors (or ARBs), which reduce both preload and afterload, can be especially helpful. Beta blockers may also improve outcome. Drug therapy of heart failure is discussed in Chapter 48. Cardiac Rupture Weakening of the myocardium predisposes the heart wall to rupture. Following rupture, shock and circulatory collapse develop rapidly. Death is often immediate. Fortunately, cardiac rupture is relatively rare (less than 2% incidence). Patients at highest risk are those with a large anterior infarction. Cardiac rupture is most likely within the first days after MI. Early treatment with vasodilators and beta blockers may reduce the risk of wall rupture. SECONDARY PREVENTION OF STEMI As a rule, patients who survive the acute phase of STEMI can be discharged from the hospital as early as 72 hours after admission if they remain free of complications. However, they are still at risk of reinfarction (5% to 15% incidence within the first year) and other complications (e.g., dysrhythmias, heart failure). Outcome can be improved with risk factor reduction, exercise, and long-term therapy with drugs. Reduction of risk factors for MI can increase long-term survival. Patients who smoke must be encouraged to quit; the goal is total cessation. Patients with high serum cholesterol should be given an appropriate dietary plan and, if necessary, treated with a high-dose statin. Hypertension and diabetes increase the risk of mortality and must be controlled. For patients with hypertension, blood pressure should be decreased to below 140/90 mm Hg. For patients with diabetes, the goal is a level of hemoglobin A1C below 7%. Exercise training can be valuable for two reasons: it reduces complications associated with prolonged bed rest and it accelerates return to an optimal level of functioning. The goal is 30 minutes of exercise at least 3 to 4 days a week. Although exercise is safe for most patients, there is concern about cardiac risk and impairment of infarct healing in patients whose infarct is large. All post-MI patients should take four drugs: (1) a beta blocker; (2) an ACE inhibitor or an ARB; either (3a) an antiplatelet drug (aspirin or clopidogrel, ticagrelor, or prasugrel) or (3b) an anticoagulant (warfarin); and (4) a statin. All four should be taken indefinitely. Estrogen therapy for postmenopausal women is not effective as secondary prevention and should not be initiated. KEY POINTS ■ ■ MI is necrosis of the myocardium secondary to acute occlusion of a coronary artery. The usual cause is platelet plugging and thrombus formation at the site of a ruptured atherosclerotic plaque. STEMI is diagnosed by the presence of chest pain, characteristic ECG changes, and elevated serum levels of cardiac troponins. ■ 638 Aspirin suppresses platelet aggregation, decreasing mortality, reinfarction, and stroke. All patients should chew a 162- to 325-mg dose on hospital admission and should take 81 to 162 mg/day indefinitely after discharge. In patients undergoing acute STEMI, beta blockers reduce cardiac pain, infarct size, short-term mortality, recurrent ischemia, and reinfarction. Continued use increases CHAPTER 53 ■ ■ ■ ■ ■ ■ ■ ■ long-term survival. All patients should receive a beta blocker in the absence of specific contraindications. Oxygen, morphine, and nitroglycerin are considered routine therapy for suspected STEMI. They should be started, as appropriate, soon after symptom onset. Reperfusion therapy, which restores blood flow through blocked coronary arteries, is the most beneficial treatment for STEMI. Reperfusion can be accomplished with PCI or with fibrinolytic drugs. Both approaches are highly effective, but PCI is now generally preferred. PCI usually consists of balloon angioplasty coupled with placement of a drug-eluting stent. Fibrinolytic drugs dissolve clots by converting plasminogen into plasmin, an enzyme that digests the fibrin meshwork that holds clots together. Typically, all fibrinolytic drugs are equally effective. However, when treatment is initiated within 4 to 6 hours of pain onset, alteplase is most effective. The major complication of fibrinolytic therapy is bleeding. Intracranial hemorrhage is the greatest concern. Heparin is recommended for all patients undergoing fibrinolytic therapy or PCI. ■ ■ ■ ■ Management of ST-Elevation Myocardial Infarction Aspirin (an antiplatelet drug) combined with clopidogrel is recommended for all patients undergoing reperfusion therapy with a fibrinolytic drug. Glycoprotein IIb/IIIa inhibitors (e.g., abciximab) are powerful IV antiplatelet drugs that can enhance the benefits of primary PCI. In patients with acute MI, ACE inhibitors decrease mortality, severe heart failure, and recurrent MI. All patients should receive an ACE inhibitor in the absence of specific contraindications. For patients who cannot tolerate ACE inhibitors, an ARB may be used instead. To lower the risk of a second MI, all patients should decrease cardiovascular risk factors (e.g., smoking, hypercholesterolemia, hypertension, diabetes), exercise for 30 minutes at least 3 or 4 days a week, and undergo long-term therapy with four drugs: a beta blocker, an ACE inhibitor or an ARB, an antiplatelet drug or warfarin, and a statin. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. 639 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ ■ Hemophilia is a bleeding disorder seen almost exclusively in males. The underlying cause is a genetically based deficiency of clotting factors. Hemophilia has two forms: hemophilia A (factor VIII deficiency) and hemophilia B (factor IX deficiency). Hemophilia may be severe, moderate, or mild, depending on the degree of clotting factor deficiency. Patients with severe hemophilia may experience lifethreatening hemorrhage in response to minor trauma, whereas those with mild hemophilia may experience little or no excessive bleeding. Repeated bleeding in the knee and other joints can cause permanent joint damage. The cornerstone of hemophilia treatment is replacement therapy with factor VIII (hemophilia A) or factor IX (hemophilia B). Replacement therapy may be done prophylactically (to prevent bleeding and thus prevent joint injury) or on demand (to stop an ongoing bleed or to prevent excessive bleeding during surgery). Clotting factor products are made in two basic ways: extraction from donor plasma and production in cell culture using recombinant DNA technology. All clotting factor concentrates, whether plasma derived or recombinant, are equally effective. All clotting factor concentrates in use today are very safe: They carry no risk of transmitting HIV/AIDS and little or no risk of transmitting hepatis or CJD. However, because recombinant factors are, in theory, slightly safer than plasma-derived factors, recombinant factors are considered the treatment of choice. As a rule, clotting factors are given by slow IV push. Continuous infusion may also be done, but only by a clinician with special training. Clotting factor dosage depends primarily on the site and severity of the bleed. A dose of 1 unit of factor VIII/kg will raise the plasma level of factor VIII activity by 2%, whereas 1 unit of factor IX/kg will raise the plasma level of factor IX activity by only 1%. ■ ■ ■ ■ ■ ■ ■ ■ ■ Although we can monitor the activity of clotting factors in blood to help guide treatment, dosage is ultimately determined by the clinical response. For prophylaxis, clotting factor concentrates are administered on a regular schedule, usually every other day to 3 times a week for factor VIII and twice a week for factor IX. With both factors, the goal is to maintain plasma factor levels above 1% of normal. To facilitate frequent IV administration during prophylaxis, a central venous access device can be installed. Clotting factor concentrates can cause allergic reactions. Mild reactions can be managed with an antihistamine (e.g., diphenhydramine [Benadryl]). The most severe reaction— anaphylaxis—is treated with subQ epinephrine. For some patients with mild hemophilia A, bleeding can be stopped with desmopressin, a drug that promotes the release of stored factor VIII. Desmopressin does not release factor IX, and so cannot treat hemophilia B. Two drugs—aminocaproic acid and tranexamic acid—can suppress fibrinolysis and can promote hemostasis in hemophilia A and hemophilia B. These antifibrinolytic agents are more effective for preventing recurrent bleeding than for stopping an ongoing bleed. Development of inhibitors (antibodies that neutralize factor VIII or factor IX) is a serious complication of hemophilia therapy. Activated factor VII (factor VIIa) and AICC are preferred agents for controlling bleeding when inhibitors of factor VIII or factor IX are present. People with hemophilia should avoid aspirin and other traditional NSAIDs because these agents suppress platelet aggregation and promote GI ulceration and bleeding. Second-generation NSAIDs (COX-2 inhibitors) are probably safe. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa FACTOR VIII AND FACTOR IX CONCENTRATES Preadministration Assessment Therapeutic Goal Factor VIII is indicated for replacement therapy in patients with hemophilia A, and factor IX is indicated for replacement therapy in patients with hemophilia B. Both factors may be given prophylactically (to prevent bleeding and subsequent joint injury) or “on demand” (to stop ongoing bleeding or prevent excessive bleeding during anticipated surgery). Baseline Data Obtain a baseline level for activity of factor VIII or factor IX. Identifying High-Risk Patients Use with caution in patients with a history of allergic reactions to the factor concentrate. 646 CHAPTER 54 Drugs for Hemophilia Summary of Major Nursing Implicationsa—cont’d therapy). With both forms of therapy, monitoring activity levels of factor VIII or factor IX can help guide treatment. Implementation: Administration Route Intravenous. Administration Administer by slow IV push or continuous infusion. Record the following each time you give a factor concentrate: • • • • Time and date Infusion site and rate Total dose Manufacturer, brand name, lot number, and expiration date of the factor concentrate Teach home caregivers about: • The importance of having an assistant, who can give aid • • • • • • or call for help if complications arise The importance and proper method of hand washing Making dosage calculations Reconstituting the powdered factor concentrate Infusion technique Cleanup and waste disposal Recording the time, date, and other information listed in this section Minimizing Adverse Effects Allergic Reactions. Clotting factor concentrates can cause allergic reactions, ranging from mild to severe. Inform patients about symptoms of mild reactions (e.g., hives, rash, urticaria, stuffy nose, fever), and advise them to take an antihistamine (e.g., diphenhydramine) if these occur. Inform patients about symptoms of anaphylaxis (wheezing, tightness in the throat, shortness of breath, swelling in the face), and instruct them to seek immediate emergency care if these develop. The treatment of choice is epinephrine, injected subQ. Minimizing Adverse Interactions Aspirin. Warn patients not to use aspirin, a drug that inhibits platelet aggregation and can cause GI ulceration and bleeding. NSAIDs Other Than Aspirin. Advise patients that firstgeneration NSAIDs (e.g., ibuprofen, naproxen) have actions similar to those of aspirin, and hence should be avoided. Advise patients that second-generation NSAIDs (e.g., celecoxib), which do not inhibit platelets and cause minimal GI effects, are probably safe. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Success is indicated by preventing bleeding (during prophylactic therapy) or controlling bleeding (during on-demand Patient education information is highlighted as blue text. a 647 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ The principal cause of iron deficiency is increased iron demand secondary to (1) maternal and fetal blood volume expansion during pregnancy; (2) blood volume expansion during infancy and early childhood; or (3) chronic blood loss, usually of GI or uterine origin. The major consequence of iron deficiency is microcytic, hypochromic anemia. Ferrous sulfate, given PO, is the drug of choice for iron deficiency. Iron-deficient patients who cannot tolerate or absorb oral ferrous salts are treated with parenteral iron—usually iron dextran administered IV. The major adverse effects of ferrous sulfate are GI disturbances. These are best managed by reducing the dosage (rather than by administering the drug with food, which would greatly reduce absorption). Parenteral iron dextran carries a significant risk of fatal anaphylactic reactions. The risk is much lower with other parenteral iron products (e.g., iron sucrose). When iron dextran is used, a small test dose is required before each full dose. Be aware, however, that patients can experience anaphylaxis and other hypersensitivity reactions from the test dose, and patients who did not react to the test dose may still have these reactions with the full dose. The principal cause of vitamin B12 deficiency is impaired absorption secondary to lack of intrinsic factor. ■ ■ ■ ■ ■ ■ ■ The principal consequences of B12 deficiency are megaloblastic (macrocytic) anemia and neurologic injury. Vitamin B12 deficiency caused by malabsorption is treated lifelong with cyanocobalamin. Traditional treatment consists of IM injections administered monthly. However, large oral doses administered daily are also effective, as are intranasal doses (administered weekly with Nascobal). For initial therapy of severe vitamin B12 deficiency, parenteral folic acid is given along with cyanocobalamin. When folic acid is combined with vitamin B12 to treat B12 deficiency, it is essential that the dosage of B12 be adequate because folic acid can mask continued B12 deficiency (by improving the hematologic picture), while allowing the neurologic consequences of B12 deficiency to progress. The principal causes of folic acid deficiency are poor diet (usually in patients with alcohol use disorder) and malabsorption secondary to intestinal disease. The principal consequences of folic acid deficiency are megaloblastic anemia and neural tube defects in the developing fetus. To prevent neural tube defects, all women who may become pregnant should ingest 400 to 800 mcg of supplemental folate daily, in addition to the folate they get in food. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa indicative of anemia are subnormal hemoglobin levels, subnormal hematocrit, subnormal hemosiderin in bone marrow, and the presence of microcytic, hypochromic erythrocytes. The cause of iron deficiency (e.g., pregnancy, occult bleeding, menorrhagia, inadequate diet, malabsorption) must be determined. IRON PREPARATIONS Carbonyl iron Ferric ammonium citrate Ferrous aspartate Ferrous bisglycinate Ferrous fumarate Ferrous gluconate Ferrous sulfate Ferumoxytol Heme-iron polypeptide Iron dextran Iron sucrose Polysaccharide iron complex Sodium–ferric gluconate complex (SFGC) Identifying High-Risk Patients All iron preparations are contraindicated for patients with anemias other than iron deficiency anemia. Parenteral preparations are contraindicated for patients who have had a severe allergic reaction to them in the past. Use oral preparations with caution in patients with peptic ulcer disease, regional enteritis, and ulcerative colitis. Except where indicated, the implications summarized here apply to all iron preparations. Preadministration Assessment Therapeutic Goal Prevention or treatment of iron deficiency anemias. Baseline Data Before treatment, assess the degree of anemia. Fatigue, listlessness, and pallor indicate mild anemia; dyspnea, tachycardia, and angina suggest severe anemia. Laboratory findings Implementation: Administration Routes Oral. Ferrous sulfate, ferrous fumarate, ferrous gluconate, ferrous aspartate, ferrous bisglycinate, ferric ammonium citrate, carbonyl iron, heme-iron polypeptide, polysaccharide iron complex, SFGC. Parenteral. Iron dextran, SFGC, iron sucrose, ferumoxytol. Oral Administration Food reduces GI distress from oral iron but also greatly reduces absorption. Instruct patients to administer oral iron between 660 CHAPTER 55 Drugs for Deficiency Anemias Summary of Major Nursing Implicationsa—cont’d meals to maximize uptake. If GI distress is intolerable, the dosage may be reduced. If absolutely necessary, oral iron may be administered with meals. Liquid preparations can stain the teeth. Instruct patients to dilute liquid preparations with juice or water, administer them through a straw, and rinse the mouth after. Warn patients not to crush or chew sustained-release preparations. Warn patients against ingesting iron salts together with antacids or tetracyclines. Inform patients that oral iron preparations differ and warn them against switching from one to another. Parenteral Administration: Iron Dextran Iron dextran may be given IV or IM. Intravenous administration is safer and preferred. Intravenous. To minimize anaphylactic reactions, follow this protocol: (1) Infuse 25 mg as a test dose and observe the patient for at least 15 minutes. (2) If the test dose appears safe, infuse 100 mg over 10 to 15 minutes. (3) If the 100-mg dose proves uneventful, give additional doses as needed every 24 hours. Intramuscular. Intramuscular injection can cause significant adverse reactions (anaphylaxis, persistent pain, localized discoloration, promotion of tumors) and is generally avoided. Make injections deep into each buttock using the Z-track technique. Give a 25-mg test dose and wait 1 hour before giving the full therapeutic dose. Parenteral Administration: SFGC To minimize adverse reactions, precede the first full dose with a test dose (25 mg infused IV over 60 minutes). Administer therapeutic doses by slow IV infusion (no faster than 12.5 mg/min). Parenteral Administration: Iron Sucrose Hemodialysis-Dependent Patients. Administer iron sucrose directly into the dialysis line. Do not mix with other drugs or with parenteral nutrition solutions. Administer by either (1) slow injection (1 mL/min) or (2) infusion (dilute iron sucrose in up to 100 mL of 0.9% saline and infuse over 15 minutes or longer). Peritoneal Dialysis–Dependent Patients. Administer by slow infusion. Non–Dialysis-Dependent Patients. Administer by slow injection. Parenteral Administration: Ferumoxytol Give 510 mg by slow IV injection, defined here as 1 mL/sec (30 mg/sec), taking about 17 seconds for the total 510-mg dose. Repeat 3 to 8 days later. Implementation: Measures to Enhance Therapeutic Effects If the diet is low in iron, advise the patient to increase consumption of iron-rich foods (e.g., egg yolks, brewer’s yeast, wheat germ, muscle meats, fish, fowl). Ongoing Evaluation and Interventions Evaluating Therapeutic Responses Evaluate treatment by monitoring hematologic status. Reticulocyte counts should increase within 4 to 7 days, hemoglobin content and the hematocrit should begin to rise within 1 week, and hemoglobin levels should rise by at least 2 gm/ dL within 1 month. If these responses do not occur, evaluate the patient for adherence, persistent bleeding, inflammatory disease, and malabsorption. Minimizing Adverse Effects GI Disturbances. Forewarn patients about possible GI reactions (nausea, vomiting, constipation, diarrhea) and inform them these will diminish over time. If GI distress is severe, the dosage may be reduced, or if absolutely necessary, iron may be administered with food. Inform patients that iron will impart a harmless dark green or black color to stools. Anaphylactic Reactions. Parenteral iron dextran (and, rarely, SFGC, iron sucrose, and ferumoxytol) can cause potentially fatal anaphylaxis. Before giving parenteral iron, ensure that injectable epinephrine and facilities for resuscitation are immediately available. After administration, observe the patient for 60 minutes. Give test doses as described earlier. Precede all doses of iron dextran with a test dose; test doses are unnecessary with iron sucrose and ferumoxytol. Managing Acute Toxicity. Iron poisoning can be fatal to young children. Instruct parents to store iron out of reach and in childproof containers. If poisoning occurs, rapid treatment is imperative. Use gastric lavage to remove iron from the stomach. Administer deferoxamine if plasma levels of iron exceed 500 mcg/mL. Manage acidosis and shock as required. CYANOCOBALAMIN (VITAMIN B12) Preadministration Assessment Therapeutic Goal Correction of megaloblastic anemia and other sequelae of vitamin B12 deficiency. Baseline Data Assess the extent of vitamin B12 deficiency. Record signs and symptoms of anemia (e.g., pallor, dyspnea, palpitations, fatigue). Determine the extent of neurologic damage. Assess GI involvement. Baseline laboratory data include plasma vitamin B12 levels, erythrocyte and reticulocyte counts, and hemoglobin and hematocrit values. Bone marrow may be examined for megaloblasts. A Schilling test may be ordered to assess vitamin B12 absorption. Identifying High-Risk Patients Use with caution in patients receiving folic acid. Implementation: Administration Routes and Administration Administration may be IM, subQ, oral, or intranasal. For most patients, lifelong treatment is required. Traditional therapy Continued 661 UNIT VII Drugs That Affect the Heart, Blood Vessels, and Blood Summary of Major Nursing Implicationsa—cont’d consists of IM or subQ injections administered monthly. However, treatment can be just as effective with large daily oral doses or with intranasal doses (administered weekly with Nascobal). Inform patients that intranasal doses should severe megaloblastic anemia resulting from vitamin B12 deficiency; and (3) prevention of folic acid deficiency, especially in women who might become pregnant and in women who are pregnant or lactating. Implementation: Measures to Enhance Therapeutic Effects Promoting Adherence Patients with permanent impairment of B12 absorption require lifelong B12 therapy. To promote adherence, educate patients Baseline Data Assess the extent of folate deficiency. Record signs and symptoms of anemia (e.g., pallor, dyspnea, palpitations, fatigue). Determine the extent of GI damage. Baseline laboratory data include serum folate levels, erythrocyte and reticulocyte counts, and hemoglobin and hematocrit values. In addition, bone marrow may be evaluated for megaloblasts. To rule out vitamin B12 deficiency, vitamin B12 determinations and a Schilling test may be ordered. not be administered within 1 hour before or 1 hour after consuming hot foods or hot liquids. about the nature of their condition, and impress upon them the need for monthly injections, daily oral therapy, or weekly intranasal therapy. Schedule appointments for injections at convenient times. Improving Nutrition When B12 deficiency is not due to impaired absorption, a change in diet may accelerate recovery. Advise the patient to increase consumption of B12-rich foods (e.g., muscle meats, dairy products). Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Assess for improvements in hematologic and neurologic status. Over a period of 2 to 3 weeks, megaloblasts should disappear, reticulocyte counts should rise, and the hematocrit should normalize. Neurologic damage may take months to improve; in some cases, full recovery may never occur. For patients receiving long-term therapy, vitamin B12 levels should be measured every 3 to 6 months, and blood counts should be performed. Minimizing Adverse Effects Hypokalemia may develop during the first days of therapy. Monitor serum potassium levels and observe the patient for signs of potassium insufficiency. Teach patients the signs and symptoms of hypokalemia (e.g., muscle weakness, irregular heartbeat), and instruct them to report these immediately. Minimizing Adverse Interactions Folic acid can correct hematologic effects of vitamin B12 deficiency, but not the neurologic effects. By improving the hematologic picture, folic acid can mask ongoing B12 deficiency, resulting in undertreatment and progression of neurologic injury. Accordingly, when folic acid and cyanocobalamin are used concurrently, special care must be taken to ensure that the cyanocobalamin dosage is adequate. FOLIC ACID (FOLACIN, FOLATE, PTEROYLGLUTAMIC ACID) Identifying High-Risk Patients Folic acid is contraindicated for patients with pernicious anemia (except during the acute phase of treatment). Inappropriate use of folic acid by these patients can mask signs of vitamin B12 deficiency, allowing further neurologic deterioration. Implementation: Dosage and Administration Routes Oral, subQ, IV, and IM. Oral administration is most common and preferred. Injections are employed only when intestinal absorption is severely impaired. Dosage Prevention of Neural Tube Defects. To reduce the risk of neural tube defects, women who might become pregnant should consume 400 to 800 mcg of supplemental folate daily—in addition to the folate they get from food. Treatment of Folate-Deficient Megaloblastic Anemia. The initial oral dosage is 1000 to 2000 mcg/day. Once symptoms have resolved, the maintenance dosage is 400 mcg/day. Implementation: Measures to Enhance Therapeutic Effects Improving Nutrition If the diet is deficient in folic acid, advise the patient to increase consumption of folate-rich foods (e.g., green vegetables, liver). If alcoholism underlies dietary deficiency, offer counseling for alcoholism, as well as dietary advice. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects Monitor hematologic status. Within 2 weeks, megaloblasts should disappear, reticulocyte counts should increase, and the hematocrit should begin to rise. Preadministration Assessment Therapeutic Goal Folic acid is used for (1) treatment of megaloblastic anemia resulting from folic acid deficiency; (2) initial treatment of Patient education information is highlighted as blue text. a 662 CHAPTER 56 Hematopoietic Agents KEY POINTS ■ ■ ■ ■ ■ ■ ■ ■ Epoetin is given to increase red blood cell counts and thereby decrease the need for transfusions. Specific indications include anemia associated with (1) chronic renal failure, (2) myelosuppressive cancer chemotherapy, and (3) zidovudine therapy in patients with HIV/AIDS. By increasing the hematocrit, epoetin can cause or exacerbate hypertension. Epoetin increases the risk of cardiovascular events (e.g., cardiac arrest, stroke, HF, MI), especially when the hemoglobin level exceeds 11 gm/dL or the rate of rise in hemoglobin exceeds 1 gm/dL in 2 weeks. In some cancer patients, epoetin can accelerate tumor progression and shorten life. Owing to the risk of serious toxicity, epoetin must be prescribed and used under a new Risk Evaluation and Mitigation Strategy (REMS). Filgrastim is given to elevate neutrophil counts and thereby reduce the risk of infection. Specific indications are chronic severe neutropenia and neutropenia associated with cancer chemotherapy or BMT. The principal adverse effects of filgrastim are bone pain and leukocytosis. Sargramostim is used to accelerate recovery from BMT, treat patients in whom a bone marrow transplant has failed, ■ ■ ■ ■ ■ ■ and accelerate neutrophil recovery in patients undergoing chemotherapy for AML. The principal adverse effect of sargramostim is leukocytosis. Oprelvekin is given to stimulate platelet production in patients undergoing myelosuppressive chemotherapy for nonmyeloid cancers. The goal is to minimize thrombocytopenia and platelet transfusions. The principal adverse effects of oprelvekin are fluid retention (which causes edema and anemia), cardiac dysrhythmias (tachycardia, atrial fibrillation, and atrial flutter), and severe allergic reactions, including anaphylaxis. TRAs are used to increase platelet production in patients with ITP after traditional methods of treatment have failed. Uncommon but serious effects of TRAs include bone marrow fibrosis, hematologic malignancy, and thrombotic/ thromboembolic complications. Since epoetin alfa, filgrastim, sargramostim, and oprelvekin stimulate proliferation of bone marrow cells, these drugs should be used with great caution, if at all, in patients with cancers of bone marrow origin. Please visit http://evolve.elsevier.com/Lehne for chapterspecific NCLEX® examination review questions. Summary of Major Nursing Implicationsa EPOETIN ALFA (ERYTHROPOIETIN) Preadministration Assessment Therapeutic Goal Epoetin is used to restore and maintain erythrocyte counts, and thereby decrease the need for transfusions, in patients with CRF, HIV-infected patients receiving zidovudine, anemic patients facing elective surgery, and cancer patients receiving myelosuppressive chemotherapy, but only if the goal of chemotherapy is palliation, not cure. For most patients, the hemoglobin level should not exceed 10 or 11 mg/dL. Implementation: Administration Routes IV and subQ. Handling and Storage Epoetin alfa is supplied in single-use and multiuse vials; don’t re-enter the single-use vials. Don’t agitate. Don’t mix with other drugs. Discard the unused portion of the vial. Store at 2°C to 8°C (36°F to 46°F); don’t freeze. Administration Chronic Renal Failure. Administer by IV bolus or subQ Baseline Data All Patients. Obtain blood pressure; blood chemistry (BUN, uric acid, creatinine, phosphorus, potassium); complete blood counts with differential and platelet count; hemoglobin level; degree of transferrin saturation (should be at least 20%); and ferritin concentration (should be at least 100 ng/mL). HIV-Infected Patients. Obtain an erythropoietin level. If the level is above 500 milliunits/mL, epoetin is unlikely to help. Identifying High-Risk Patients Avoid epoetin alfa in patients with uncontrolled hypertension, hypersensitivity to mammalian cell–derived products or albumin, or cancer of myeloid origin. injection. Chemotherapy-Induced Anemia. Administer by subQ injection. Zidovudine-Induced Anemia. Administer by IV or subQ injection. Surgery Patients. Administer by subQ injection. Ongoing Evaluation and Interventions Monitoring Summary Measure hemoglobin level twice weekly until the maximum acceptable level has been achieved (10 or 11 mg/dL for most patients) and a maintenance dosage established. Measure hemoglobin periodically thereafter. Obtain complete blood counts with a differential and platelet counts routinely. Monitor Continued 671 Summary of Major Nursing Implicationsa—cont’d blood chemistry, including BUN, uric acid, creatinine, phosphorus, and potassium. Monitor iron stores and maintain at an adequate level. Monitor blood pressure. Minimizing Adverse Effects Hypertension. Monitor blood pressure and, if necessary, control with antihypertensive drugs. If hypertension cannot be controlled, reduce epoetin dosage. In patients with preexisting hypertension (a common complication of CRF), make certain that blood pressure is controlled before epoetin use. Cardiovascular Events. Epoetin has been associated with an increase in cardiovascular events (e.g., cardiac arrest, stroke, HF, and MI). Risk is greatest when the hemoglobin level exceeds 11 gm/dL or the rate of rise in hemoglobin exceeds 1 gm/dL in 2 weeks. To minimize risk, reduce dosage when hemoglobin approaches 11 gm/dL or when the rate of rise exceeds 1 gm/dL in 2 weeks, and temporarily stop dosing if hemoglobin rises to 11 gm/dL or more. CRF patients on dialysis may need a higher dosage of heparin to prevent clotting in the dialysis machine. For patients taking the drug before elective surgery, anticoagulant treatment can reduce the risk of deep vein thrombosis. Cancer Patients: Tumor Progression and Shortened Survival. Epoetin can accelerate tumor progression and shorten survival in some cancer patients. To reduce risk, dosage should be no higher than needed to bring hemoglobin gradually up to 12 gm/dL. Also, epoetin should be used only in cancer patients who are undergoing chemotherapy or radiation therapy. Those who are not receiving chemotherapy or radiation therapy should not take this drug. Autoimmune Pure Red-Cell Aplasia. Epoetin use may lead to pure red-cell aplasia (PRCA), owing to production of neutralizing antibodies directed against epoetin and native erythropoietin. If evidence of PRCA develops, epoetin should be discontinued and blood assessed for neutralizing antibodies. If PRCA is diagnosed, transfusions will be needed for life. Patient Education. Give all patients a Medication Guide that explains the risks and benefits of epoetin so that they can make an informed decision on whether to use this drug. FILGRASTIM (GRANULOCYTE COLONY-STIMULATING FACTOR) Preadministration Assessment Therapeutic Goal Filgrastim is given to promote neutrophil recovery in cancer patients following myelosuppressive chemotherapy or BMT. The drug is also used to treat severe chronic neutropenia. Baseline Data Obtain complete blood counts and platelet counts. Identifying High-Risk Patients Filgrastim is contraindicated for patients with hypersensitivity to Escherichia coli–derived proteins. Use with caution in patients with cancers of bone marrow origin. Implementation: Administration Routes IV, subQ. Handling and Storage Filgrastim is supplied in single-use vials. Don’t re-enter the vial; discard the unused portion. Don’t agitate. Store at 2°C to 8°C (36°F to 46°F); don’t freeze. Before administration, filgrastim may be kept at room temperature for up to 24 hours. Administration Cancer Chemotherapy. Administer by subQ bolus, short IV infusion, or continuous IV or subQ infusion. Bone Marrow Transplantation. Administer by slow IV or subQ infusion. Chronic Severe Neutropenia. Inject subQ daily. Ongoing Evaluation and Interventions Evaluating Therapeutic Effects. Obtain complete blood counts twice weekly. Discontinue treatment when the absolute neutrophil count reaches 10,000/mm3. Minimizing Adverse Effects Bone Pain. Evaluate for bone pain and treat with a nonopioid analgesic (e.g., acetaminophen). Consider an opioid analgesic if the nonopioid is insufficient. Leukocytosis. Massive doses can cause leukocytosis (white blood cell counts above 100,000/mm3). If leukocytosis develops, reduce filgrastim dosage. SARGRAMOSTIM (GRANULOCYTE-MACROPHAGE COLONY-STIMULATING FACTOR) Preadministration Assessment Therapeutic Goal Sargramostim is used to accelerate myeloid recovery in cancer patients who have undergone autologous BMT following high-dose chemotherapy (with or without concurrent irradiation). In addition, the drug is approved for treatment of patients for whom an autologous or allogenic bone marrow transplant has failed to take. Sargramostim is also used to accelerate neutrophil recovery in older patients receiving induction chemotherapy for AML. Baseline Data Obtain complete blood counts with differential and platelet count. Identifying High-Risk Patients Sargramostim is contraindicated in the presence of hypersensitivity to yeast-derived products and excessive leukemic myeloid blasts in bone marrow or peripheral blood. Exercise caution in patients with cardiac disease, hypoxia, peripheral edema, pleural or pericardial effusion, or cancers of bone marrow origin. Implementation: Administration Route IV (by infusion). Handling and Storage Sargramostim is supplied in concentrated solution and as a powder, which must be reconstituted for IV infusion. To CHAPTER 56 Hematopoietic Agents Summary of Major Nursing Implicationsa—cont’d reconstitute the powder, add 1 mL of sterile water and gently swirl. Before infusing, dilute the concentrated solution or reconstituted powder. Administer as soon as possible after diluting—and no later than 6 hours after reconstitution. Store sargramostim (powder, reconstituted powder, final IV solution) at 2°C to 8°C (36°F to 46°F) until used. Administration Administer by 2-hour or 4-hour IV infusion. Ongoing Evaluation and Interventions Minimizing Adverse Effects Leukocytosis and Thrombocytosis. Obtain complete blood counts with differential and platelet counts twice weekly. If the white blood cell count rises above 50,000/mm3, if the absolute neutrophil count rises above 20,000/mm3, or if the platelet count rises above 500,000/mm3, temporarily interrupt sargramostim or reduce the dosage. OPRELVEKIN (INTERLEUKIN-11) Preadministration Assessment Therapeutic Goal Oprelvekin is given to minimize thrombocytopenia and the need for platelet transfusions in patients undergoing myelosuppressive therapy for nonmyeloid cancers. Baseline Data Determine baseline blood cell counts and platelet count, hematocrit, and fluid and electrolyte status. Identifying High-Risk Patients Use with caution in patients with cancers of myeloid origin; in patients taking diuretics or ifosfamide; and in patients with a history of atrial dysrhythmias, HF, pleural effusion, or papilledema. Implementation: Administration Route SubQ. Handling and Storage Oprelvekin is supplied in single-use vials; don’t re-enter the vial. Don’t agitate. Don’t mix with other drugs. Discard the unused portion of the vial. Store at 2°C to 8°C (36°F to 46°F); don’t freeze. Administration Administer once daily beginning 4 to 6 hours after chemotherapy. Continue for 21 days or until platelet counts exceed 50,000/mm3—whichever comes first. Ongoing Evaluation and Interventions Monitoring Summary Monitor platelet counts from the time of the expected nadir until the count exceeds 50,000/mm3. Monitor blood cell counts, fluid status, and electrolyte status. Minimizing Adverse Effects Fluid Retention. Fluid retention can result in edema, expanded plasma volume, anemia, and dyspnea. Instruct patients with a history of congestive heart failure or pleural effusion to contact the prescriber if dyspnea worsens. Cardiac Dysrhythmias. Oprelvekin can cause tachycardia, atrial flutter, and atrial fibrillation. Use caution in patients with a history of these disorders. ROMIPLOSTIM Preadministration Assessment Therapeutic Goal Romiplostim is given to increase platelet production in patients with ITP that has not responded to other conventional treatments. Baseline Data Determine baseline blood cell counts and platelet count. Identifying High-Risk Patients Use with caution in patients with cancers of myeloid origin, patients with hematologic malignancies, and patients with hepatic or renal impairment. Implementation: Administration Route SubQ. Handling and Storage Romiplostim is supplied in single-use vials; don’t re-enter the vial. Don’t agitate. Protect the reconstituted medication from light. Don’t mix with other drugs. Discard the unused portion of the vial. Store at 2°C to 8°C (36°F to 46°F); don’t freeze. Administration Administer 1 mcg/kg once weekly and adjust dose based on platelet response. Use lowest effective dose to maintain platelets above 50,000/mm3. Ongoing Evaluation and Interventions Monitoring Summary Monitor platelet counts from the time of the expected nadir until the count exceeds 50,000/mm3. Monitor blood cell counts weekly until platelet counts are stable for 4 weeks. Then monitor platelets and blood count every 2 months thereafter. Minimizing Adverse Effects Thrombosis/Thromboembolism. Romiplostim should not be used to normalize platelet counts. Depending on current platelet count, doses should be adjusted per package recommendations. In patients with chronic liver disease, portal vein thrombosis has been reported with romiplostim use. Use cautiously in this population of patients. Patient education information is highlighted as blue text. a 673 CHAPTER 76 Drugs for Asthma and Chronic Obstructive Pulmonary Disease Tobacco smoke Air pollution Inherited 1-antitrypsin deficiency Inflammation of the airway epithelium Infiltration of inflammatory cells and release of cytokines (neutrophils, macrophages, lymphocytes, leukotrienes, interleukins) Systemic effects (muscle weakness, weight loss) Inhibition of normal endogenous antiproteases Continuous bronchial irritation and inflammation Increased protease activity with breakdown of elastin in connective tissue of lungs (elastases, cathepsins, etc.) Chronic bronchitis (bronchial edema, hypersecretion of mucus, bacterial colonization of airways) Emphysema (destruction of alveolar septa and loss of elastic recoil of bronchial walls) Airway obstruction Air trapping Loss of surface area for gas exchange Frequent exacerbations (infections, bronchospasm) Dyspnea Cough Hypoxemia Hypercapnia Cor pulmonale Fig. 76.2 ■ Pathogenesis of chronic bronchitis and emphysema. Prototype Drugs DRUGS FOR ASTHMA AND COPD Anti-Inflammatory Drugs: Glucocorticoids Beclomethasone (inhaled) Prednisone (oral) Anti-Inflammatory Drugs: Others Cromolyn (mast cell stabilizer, inhaled) Zafirlukast (leukotriene modifier, oral) Bronchodilators: Methylxanthines Theophylline Anticholinergic Drugs Ipratropium Bronchodilators: Beta2-Adrenergic Agonists Albuterol (inhaled, short acting) Salmeterol (inhaled, long acting) 927 UNIT XIII Respiratory Tract Drugs TABLE 76.1 ■ Overview of Major Drugs for Asthma and Chronic Obstructive Pulmonary Disease ANTI-INFLAMMATORY DRUGS Glucocorticoids BRONCHODILATORS Beta2-Adrenergic Agonists Inhaled Inhaled: Short Acting Beclomethasone dipropionate [QVAR] Budesonide [Pulmicort Flexhaler, Pulmicort Respules, Pulmicort Turbuhaler ] Ciclesonide [Alvesco] Flunisolide [Aerospan] Fluticasone propionate [Flovent HFA, Flovent Diskus] Mometasone furoate [Asmanex Twisthaler] Albuterol [ProAir HFA, ProAir RespiClick, Proventil HFA, Ventolin HFA, Airomir, Apo-Salvent MDI ] Levalbuterol [Xopenex, Xopenex HFA] Inhaled: Long Actingc Arformoterol [Brovana]b Formoterol [Foradil Aerolizer, Perforomist, Oxeze Turbuhaler Indacaterol [Arcapta Neohaler, Onbrez Breezhaler ]b Olodaterol [Striverdi Respimat]b Salmeterol [Serevent Diskus]c Oral Methylprednisolone [A-Methapred, Depo-Medrol, Medrol, Medrol Dose-Pak] Prednisolone [Flo-Pred, Orapred ODT, Millipred, Pediapred, Prelone] Prednisone [Deltasone, Winpred ] Oral Albuterol [VoSpire ER] Terbutaline (generic only) Methylxanthines Leukotriene Modifiers Aminophylline, oral (generic only) Theophylline, oral [Theo-24, Elixophyllin, Theochron, Theolair Pulmophylline , Theo ER , Uniphyl ] Montelukast, oral [Singulair] Zafirlukast, oral [Accolate]a Zileuton, oral [Zyflo, Zyflo CR]a a Aclidinium bromide, inhaled [Tudorza Pressair]b Glycopyrronium bromide, inhaled [Seebri Neohaler, Seebri Breezhaler ]b Ipratropium, inhaled [Atrovent HFA] Tiotropium, inhaled [Spiriva, Spiriva HandiHaler, Spiriva Respimat]b Umeclidinium, inhaled [Incruse Ellipta] ]a IgE Antagonist Omalizumab, subQ [Xolair] Phosphodiesterase-4 Inhibitors Roflumilast, oral [Daliresp, Daxas , Anticholinergics Cromolyn Cromolyn, inhaled [Nalcrom ]c ]b BETA AGONIST/CHOLINERGIC ANTAGONIST COMBINATIONS ANTI-INFLAMMATORY/BRONCHODILATOR COMBINATIONS Budesonide/formoterol, inhaled [Symbicort] Fluticasone/salmeterol, inhaled [Advair Diskus, Advair HFA] Fluticasone/vilanterol, inhaled [Breo Ellipta] Mometasone/formoterol, inhaled [Dulera, Zenhale Albuterol/ipratropium, inhaled [Combivent Respimat, Combivent UDV ]b Indacaterol/glycopyrronium, inhaled [Utibron Neohaler, Ultibro Breezhaler ]b Olodaterol/tiotropium, inhaled [Stiolto Respimat]b Vilanterol/umeclidinium, inhaled [Anoro Ellipta]b ] Approved only for asthma, not for chronic obstructive pulmonary disease. Approved only for chronic obstructive pulmonary disease, not for asthma. c For treatment of asthma, must always be combined with an inhaled glucocorticoid. a b ADMINISTERING DRUGS BY INHALATION Most antiasthma drugs can be administered by inhalation. This route has three advantages: (1) therapeutic effects are enhanced by delivering drugs directly to their site of action, (2) systemic effects are minimized, and (3) relief of acute attacks is rapid. Four types of inhalation devices are employed: metered-dose inhalers, Respimats, dry-powder inhalers, and nebulizers. Metered-Dose Inhalers Metered-dose inhalers (MDIs) are small hand-held, pressurized devices that deliver a measured dose of drug with each actuation. Dosing is usually accomplished with 1 or 2 inhalations. When 2 inhalations are needed, an interval of at least 1 minute should separate the first inhalation from the second. When using most MDIs, the patient must begin to inhale before activating the device. This requires hand-breath coordination, making MDIs difficult to use correctly. Accordingly, patients will need a demonstration, as well as written and verbal instruction. Even with optimal use, only about 10% of the dose reaches the lungs. About 80% affects the oropharynx and is swallowed, and the remaining 10% is left in the device or exhaled. Spacers are devices that attach directly to the MDI to increase delivery of drug to the lungs and decrease deposition of drug on the oropharyngeal mucosa (Fig. 76.3). Several kinds of spacers are available for use with MDIs. Some spacers contain a one-way valve that activates upon inhalation, obviating the need for good hand-breath coordination. Some spacers also contain an alarm whistle that sounds off when inhalation is too rapid, thus maximizing effective drug administration. They can also prevent bronchospasm that may occur with sudden intake of an inhaled drug. Respimats Respimats are inhalers that deliver drugs as a very fine mist. Like MDIs, they are activated by the user; however, the device does not use propellants. An advantage of this system is that the extremely small particle size ensures greater delivery of drug to the lungs; in addition, because less drug falls out of 928 CHAPTER 76 Drugs for Asthma and Chronic Obstructive Pulmonary Disease With Spacer Without Spacer Spacer Fig. 76.3 ■ 57% Inhaler device 10% 22% Mouth and throat 81% 21% Lungs 9% Effect of a spacer device on the distribution of inhaled medication. Note that, when a spacer is used, more medication reaches its site of action in the lungs, and less is deposited in the mouth and throat. the mist due to particle weight, there is decreased drug deposition in the mouth and oropharynx. PATIENT-CENTERED CARE ACROSS THE LIFE SPAN Anti-Inflammatory Agents Dry-Powder Inhalers Dry-powder inhalers (DPIs) are used to deliver drugs in the form of a dry micronized powder directly to the lungs. Unlike MDIs, DPIs are breath activated. As a result, DPIs don’t require the hand-breath coordination needed with MDIs, making DPIs much easier to use. Compared with MDIs, DPIs deliver more drug to the lungs (20% of the total released vs. 10%) and less to the oropharynx. Also, spacers are not used with DPIs. Life Stage Patient Care Concerns Children Inhaled glucocorticoids are the preferred long-term treatment for children of all ages, including infants. Face masks are recommended for the administration of inhaled glucocorticoids to children younger than 4 years. Alternative treatments include cromolyn and leukotriene receptor antagonists (e.g., montelukast), but evidence supporting these drugs for asthma management is lower than that supporting inhaled glucocorticoids. Montelukast is the only leukotriene modifier approved for children ages 1 to 5 years. Pregnant women Inhaled glucocorticoids are classified in FDA Pregnancy Risk Category Ca; however, they are preferred for uncontrolled asthma in pregnant women because uncontrolled asthma is associated with greater fetal risks. Of the leukotriene modifiers, montelukast and zafirlukast are Pregnancy Risk Category B,a while zileuton is Risk Category C.a Breast-feeding women Inhaled glucocorticoids are not a contraindication to breast-feeding; however, women taking systemic glucocorticoids should not breast-feed. Older adults Benefits exceed risk. Inhaled glucocorticoids are much safer for this population than systemic formulations. Nebulizers A nebulizer is a small machine used to convert a drug solution into a mist. The droplets in the mist are much finer than those produced by inhalers, resulting in less drug deposit on the oropharynx and increased delivery to the lung. Inhalation of the nebulized mist can be done through a face mask or through a mouthpiece held between the teeth. Because the mist produced by a nebulizer is inhaled with each breath, hand-breath coordination is not a concern. Nebulizers take several minutes to deliver the same amount of drug contained in 1 inhalation from an inhaler, but for some patients, a nebulizer may be more effective than an inhaler. Although nebulizers are usually used at home or in a clinic or hospital, these devices, which weigh less than 10 pounds, are sufficiently portable for use in other locations. ANTI-INFLAMMATORY DRUGS Anti-inflammatory drugs—especially inhaled glucocorticoids— are the foundation of asthma and COPD therapy. These drugs are taken daily for long-term control. Most people with asthma require these drugs for management at some point. As of 2020, the FDA will no longer use Pregnancy Risk Categories. Please refer to Chapter 9 for more information. a 929 UNIT XIII Respiratory Tract Drugs GLUCOCORTICOIDS Glucocorticoids (e.g., budesonide, fluticasone) are the most effective drugs available for long-term control of airway inflammation. Administration is usually by inhalation, but may also be IV or oral. Adverse reactions to inhaled glucocorticoids are generally minor, as are reactions to systemic glucocorticoids taken acutely. However, when systemic glucocorticoids are used long term, severe adverse effects are likely. The basic pharmacology of the glucocorticoids is presented in Chapter 72. Discussion here is limited to their use in asthma. Mechanism of Antiasthma Action Glucocorticoids reduce asthma symptoms by suppressing inflammation. Specific anti-inflammatory effects include: • Decreased synthesis and release of inflammatory mediators (e.g., leukotrienes, histamine, prostaglandins) • Decreased infiltration and activity of inflammatory cells (e.g., eosinophils, leukocytes) • Decreased edema of the airway mucosa (secondary to a decrease in vascular permeability) By suppressing inflammation, glucocorticoids reduce bronchial hyperreactivity and decrease airway mucus production. There is also some evidence that glucocorticoids may increase the number of bronchial beta2 receptors, as well as their responsiveness to beta2 agonists. Use in Asthma Glucocorticoids are used for prophylaxis of chronic asthma. Accordingly, dosing must be done on a fixed schedule—not PRN. Because beneficial effects develop slowly, these drugs cannot be used to abort an ongoing attack. Glucocorticoids do not alter the natural course of asthma, even when used in young children. Inhalation Use. Inhaled glucocorticoids are first-line therapy for management of the inflammatory component of asthma. Most patients with persistent asthma should use these drugs daily. Inhaled glucocorticoids are very effective and are much safer than systemic glucocorticoids. Oral Use. Oral glucocorticoids may be required for patients with moderate to severe persistent asthma or for management of acute exacerbations of asthma or COPD. Because of their potential for toxicity, these drugs are prescribed only when symptoms cannot be controlled with safer medications (inhaled glucocorticoids, inhaled beta2 agonists). Because the risk for toxicity increases with duration of use, treatment should be as brief as possible. low. In contrast, with prolonged use of oral glucocorticoids, adrenal suppression can be profound. Glucocorticoids can slow growth in children and adolescents—but these drugs do not decrease adult height. Short-term studies have shown that inhaled glucocorticoids slow growth; however, long-term studies indicate that adult height, while delayed, is not reduced. Less is known regarding whether glucocorticoids suppress growth and development of the brain, lungs, and other organs, in part because having asthma alone can affect organ growth. Because the benefits of inhaled glucocorticoids tend to be much greater than the risks, current guidelines for asthma management recommend these drugs for children while monitoring for evidence of complications. Long-term use of inhaled glucocorticoids may promote bone loss. Fortunately, the amount of loss is much lower than the amount caused by oral glucocorticoids. To minimize bone loss, patients should (1) use the lowest dose that controls symptoms, (2) ensure adequate intake of calcium and vitamin D, and (3) participate in weight-bearing exercise. There has been concern that prolonged therapy might increase the risk for cataracts and glaucoma. While this may be an issue of concern with continuous use of high-dose inhaled glucocorticoids, this problem is not associated with long-term use of low to medium doses of inhaled glucocorticoids. Oral Glucocorticoids. When used acutely (less than 10 days), even in very high doses, oral glucocorticoids do not cause significant adverse effects. However, prolonged therapy, even in moderate doses, can be hazardous. Potential adverse effects include adrenal suppression, osteoporosis, hyperglycemia, peptic ulcer disease, and, in young patients, growth suppression. Adrenal suppression is of particular concern. As discussed in Chapter 72, prolonged glucocorticoid use can decrease the ability of the adrenal cortex to produce glucocorticoids of its own. This can be life-threatening at times of severe physiologic stress (e.g., surgery, trauma, or systemic infection). Because Adverse Effects Inhaled Glucocorticoids. These preparations are largely devoid of serious toxicity, even when used in high doses. The most serious concern is adrenal suppression. The most common adverse effects are oropharyngeal candidiasis and dysphonia (hoarseness, speaking difficulty). Both effects result from local deposition of inhaled glucocorticoids. To minimize these effects, patients should rinse the mouth with water and gargle after each administration. Using a spacer device can help too. If candidiasis develops, it can be treated with an antifungal drug. With long-term, high-dose therapy, some adrenal suppression may develop, although the degree of suppression is generally 930 Safety Alert COMPENSATING FOR ADRENAL INSUFFICIENCY When patients have been on prolonged systemic glucocorticoid therapy, the adrenal glands decrease their endogenous production of glucocorticoids. If systemic therapy is stopped suddenly, as when switching from oral therapy to inhalation therapy, the patient can die. Similarly, during times of severe physical stress when the body would normally produce high levels of glucocorticoids, if the dose of systemic glucocorticoids is not increased to meet the increased need, the patient can die. What important lesson can you take from this? When discontinuing a systemic glucocorticoid, you must be sure that it is done gradually to allow the body to resume production of the endogenous hormone. On the other hand, if a patient taking systemic glucocorticoids experiences severe physical stress, such as a motor vehicle crash, or is scheduled for a stressful procedure such as surgery, you must make certain that the provider remembers to prescribe additional glucocorticoids to supplement for the endogenous hormone that the patient cannot produce. UNIT XIII Respiratory Tract Drugs Metabolism. Theophylline is metabolized in the liver. Rates of metabolism are affected by multiple factors—age, disease, drugs—and show wide individual variation. As a result, the plasma half-life of theophylline varies considerably among patients. For example, although the average half-life in nonsmoking adults is about 8 hours, the half-life can be as short as 2 hours in some adults and as long as 15 hours in others. Smoking either tobacco or marijuana accelerates metabolism and decreases the half-life. The average half-life in children is 4 hours. Metabolism is slowed in patients with certain pathologies (e.g., heart disease, liver disease, prolonged fever). Some drugs (e.g., cimetidine, fluoroquinolone antibiotics) decrease theophylline metabolism. Other drugs (e.g., phenobarbital) accelerate metabolism. Because of these variations in metabolism, dosage must be individualized. Drug Levels. Safe and effective therapy requires periodic measurement of theophylline blood levels. Traditionally, dosage has been adjusted to produce theophylline levels between 10 and 20 mcg/mL. However, many patients respond well at 5 mcg/mL, and, as a rule, there is little benefit to increasing levels above 15 mcg/mL. Therefore, levels between 5 and 15 mcg/mL are appropriate for most patients. At levels above 20 mcg/mL, the risk for significant adverse effects is high. the following dose should not be doubled, because doing so could produce toxicity. Smokers require higher-than-average doses. Conversely, patients with heart disease, liver dysfunction, or prolonged fever are likely to require lower doses. Patients should be instructed not to chew the sustained-release tablets or capsules. Product information should be consulted for compatibility with food. The initial dosage is based on the age and weight of the patient and on the presence or absence of factors that can impair theophylline elimination. Specific initial dosages are described in the prescribing information in the package insert. As noted previously, maintenance doses should be adjusted to produce drug levels in the therapeutic range—typically 5 to 15 mcg/mL. Intravenous. Intravenous theophylline is reserved for emergencies. Administration must be done slowly, because rapid injection can cause fatal cardiovascular reactions. Intravenous theophylline is incompatible with many other drugs. Accordingly, compatibility should be verified before mixing theophylline with other IV agents. For specific IV dosages, refer to the discussion of aminophylline that follows. Toxicity Aminophylline is a theophylline salt that is considerably more soluble than theophylline itself. In solution, each molecule of aminophylline dissociates to yield two molecules of theophylline. Hence, the pharmacologic properties of aminophylline and theophylline are identical. Aminophylline is available in formulations for oral and IV dosing. Intravenous administration is employed most often. Administration and Dosage Intravenous. Because of its relatively high solubility, aminophylline is the preferred form of theophylline for IV use. Infusions should be done slowly (no faster than 25 mg/min), because rapid injection can produce severe hypotension and death. The usual loading dose of theophylline is 4.6 mg/kg (5.7 mg/kg as aminophylline). The maintenance infusion rate should be adjusted to provide plasma levels of theophylline that are within the therapeutic range (10 to 20 mcg/mL). Aminophylline solutions are incompatible with many other drugs. Therefore, compatibility must be verified before mixing aminophylline with other IV agents. Oral. Aminophylline is available in 100- and 200-mg tablets. Dosing guidelines are the same as for theophylline. Symptoms. Toxicity is related to theophylline levels. Adverse effects are uncommon at plasma levels below 20 mcg/mL. At 20 to 25 mcg/mL, relatively mild reactions occur (e.g., nausea, vomiting, diarrhea, insomnia, restlessness). Serious adverse effects are most likely at levels above 30 mcg/ mL. These reactions include severe dysrhythmias (e.g., ventricular fibrillation) and convulsions that can be highly resistant to treatment. Death may result from cardiorespiratory collapse. Treatment. At the first indication of toxicity, dosing with theophylline should stop. Absorption can be decreased by administering activated charcoal together with a cathartic. Ventricular dysrhythmias respond to lidocaine. Intravenous diazepam may help control seizures. Drug Interactions Caffeine. Caffeine is a methylxanthine with pharmacologic properties like those of theophylline (see Chapter 36). Accordingly, caffeine can intensify the adverse effects of theophylline on the CNS and heart. In addition, caffeine can compete with theophylline for drug-metabolizing enzymes, causing theophylline levels to rise. Because of these interactions, individuals taking theophylline should avoid caffeine-containing beverages (e.g., coffee, many soft drinks) and other sources of caffeine. Tobacco and Marijuana Smoke. Smoking tobacco or marijuana can induce theophylline metabolism, resulting in increased drug clearance of up to 50% in adults and 80% in older adults. (Secondhand smoke can result in similarly decreased drug levels.) Consequently, if a smoking patient stops smoking but the dose of theophylline is not decreased, the patient is at risk for theophylline toxicity over time. Drugs That Reduce Theophylline Levels. Several agents—including phenobarbital, phenytoin, and rifampin—can lower theophylline levels by inducing hepatic drug-metabolizing enzymes. Concurrent use of these agents may necessitate an increase in theophylline dosage. Drugs That Increase Theophylline Levels. Several drugs—including cimetidine and the fluoroquinolone antibiotics (e.g., ciprofloxacin)—can elevate plasma levels of theophylline, primarily by inhibiting hepatic metabolism. To avoid theophylline toxicity, the dosage of theophylline should be reduced when the drug is combined with these agents. Formulations Theophylline is available for IV and oral use. For IV use, generic solutions are available in concentrations of 400 mg/250 mL, 400 mg/500 mL, or 800 mg/500 mL of solution. For oral use, the following concentrations are available. • • • • • • Elixophyllin oral elixir: 80 mg/15 mL Generic oral solution: 80 mg/15 mL Theochron 12-hour extended-release tablets: 100, 200, and 300 mg Generic 12-hour extended-release tablets: 100, 200, 300, and 450 mg Theo-24 24-hour extended-release capsules: 100, 200, 300, and 400 mg Generic 24-hour extended-release tablets: 400 and 600 mg Unlike the elixir and oral solution, the sustained-release tablets and capsules produce drug levels that are relatively stable. Accordingly, the sustained-release formulations are preferred for routine therapy. Dosage and Administration Oral. Dosage must be individualized. To minimize chances of toxicity, doses should be low initially and then gradually increased. If a dose is missed, Other Methylxanthines Aminophylline ANTICHOLINERGIC DRUGS Anticholinergic drugs improve lung function by blocking muscarinic receptors in the bronchi, reducing bronchoconstriction. Two agents are available: ipratropium and tiotropium. These drugs are approved only for COPD but are used off-label for asthma. Both drugs are administered by inhalation. The principal difference between the two is pharmacokinetic: Tiotropium has a much longer duration of action and thus can be dosed less often. With both drugs, systemic effects are minimal. Ipratropium Actions and Therapeutic Use Ipratropium [Atrovent HFA] is an atropine derivative administered by inhalation to relieve bronchospasm. The drug has FDA approval only for bronchospasm associated with COPD, but is often used off-label for asthma and is included in current evidence-based guidelines from the National Asthma Education and Prevention Program (NAEPP) for asthma management. Like atropine, ipratropium is a muscarinic antagonist. By blocking muscarinic cholinergic receptors in the bronchi, ipratropium prevents bronchoconstriction. Therapeutic effects begin within 30 seconds, reach 50% of their maximum in 3 minutes, and persist about 6 hours. Ipratropium is effective against allergen-induced asthma and EIB, but is less effective than the beta2 agonists. However, because ipratropium and the beta2-adrenergic agonists promote bronchodilation by different mechanisms, their beneficial effects are additive. Adverse Effects Systemic effects are minimal because ipratropium is a quaternary ammonium compound and therefore always carries a positive charge. As a result, the drug is not readily absorbed from the lungs or from the digestive tract. The most common adverse reactions are dry mouth and irritation of the pharynx. If systemic absorption is sufficient, the drug may raise intraocular pressure in patients with glaucoma. Adverse cardiovascular events (heart attack, stroke, death) have occurred in people taking ipratropium; however, because absorption is minimal, it seems unlikely that ipratropium is the cause. 938