Drugs affecting circulation: antihypertensives, antianginals, antithrombotics

Published on 12/06/2015 by admin

Filed under Pulmolory and Respiratory

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 5147 times

CHAPTER 22

Drugs affecting circulation: antihypertensives, antianginals, antithrombotics

Objectives

After reading this chapter, the reader will be able to:

1. Define terms that pertain to drugs affecting circulation: antihypertensives, antianginals, and antithrombotics

2. Categorize the stages of normal to high blood pressure

3. Define a hypertensive crisis, and differentiate between hypertensive emergency and hypertensive urgency.

4. Design an algorithm for the pharmacotherapy of hypertension.

5. Compare and contrast the clinical pharmacology of the agents used for hypertensive pharmacotherapy

6. Describe the chronotherapeutic effect of blood pressure, and design a pharmacotherapy regimen based on this principle

7. Describe the mechanism of action of angiotensin-converting enzyme inhibitors, calcium channel blockers, and β blockers

8. Compare and contrast the clinical pharmacology of spironolactone and eplerenone

9. List drug-drug interactions relevant to antihypertensives and plausible mechanisms

10. Describe the formation and elimination of an acute coronary thrombus

11. Describe the pathophysiology of angina and the drugs used to treat angina

12. List the agents in each of the following antithrombotic classes: anticoagulants, antiplatelets, and thrombolytics

13. Describe the mechanism of action of heparin

14. Compare and contrast the clinical pharmacology of heparin and low-molecular-weight heparin (LMWH)

15. List the laboratory parameters that may be used to monitor for the effect of heparin, LMWH, and direct thrombin inhibitors

16. Describe the mechanism of heparin-induced and warfarin-induced paradoxical thrombosis

17. Compare and contrast the clinical pharmacology of aspirin, clopidogrel, ticlopidine, and dipyridamole

18. Describe the role of genetic polymorphism in the antiplatelet activity of clopidogrel and anticoagulant effect of warfarin

19. Describe the indication and mechanism of action of glycoprotein IIb/IIIa inhibitors

20. List the indications and contraindication of thrombolytic agents

Key terms and definitions

Antithrombotic

Drug that prevents or breaks up blood clots in conditions such as thrombosis or embolism; antithrombotics include anticoagulants, antiplatelets, and thrombolytics.

Arterial blood pressure (blood pressure)

Defined hemodynamically as the product of systemic vascular resistance and cardiac output (heart rate × stroke volume).

Cardiovascular disease (CVD)

Damage to the heart and the blood vessels or circulation, including to the brain, kidney, and the eye.

Chronotropic

Influencing the rate of rhythmic movements (heartbeat).

Circadian rhythm

Human biologic variations of rhythm within a 24-hour cycle.

Creatinine clearance

Measurement of the renal clearance of endogenous creatinine per unit time; approximates glomerular filtration rate (GFR) but overestimates GFR by 10% to 15%; used for drug-dosing guidelines.

D-dimers

Covalently cross-linked degradation fragments of the cross-linked fibrin polymer during plasmin-mediated fibrinolysis; level increases after the onset of fibrinolysis and allows for identification of the presence of fibrinolysis.

Dose-ceiling effect

Maximum dose of a drug, beyond which it no longer exerts a therapeutic effect; however, its toxic effect increases.

Fibrin split or fibrinogen degradation products (FDPs)

Small peptides that result following the action of plasmin on fibrinogen and fibrin in the fibrinolytic process. FDPs are anticoagulant substances that can cause bleeding if fibrinolysis becomes uncontrolled and excessive.

Glomerular filtration rate (GFR)

Volume of water filtered from the plasma by the kidney via the glomerular capillary walls into Bowman capsules per unit time; considered to be 90% of creatinine clearance and equivalent to insulin clearance.

Hypertensive emergency

Blood pressure greater than 180/120 mm Hg, with the elevation of blood pressure accompanied by acute, progressing target organ injury.

Hypertensive urgency

Blood pressure greater than 180/120 mm Hg without signs or symptoms of acute target organ complications.

Inotrope

Drug influencing the contractility of a muscle (heart).

Intrinsic sympathomimetic activity (ISA)

Having the ability to activate and block adrenergic receptors, producing a net stimulatory effect on the sympathetic nervous system.

Pharmacotherapy

Treatment of disease by drug therapy.

Renin

Enzyme also known as angiotensinogenase, released by the kidney in response to a lack of renal blood flow and responsible for converting angiotensinogen into angiotensin I.

Substitute neurotransmitters

Neurotransmitter or hormone replacements that may be weaker or inert.

The circulatory system comprises an integral functional part of the cardiopulmonary system. Drug therapy affecting the circulation is seen in the acute critical care, outpatient care, and home care environments. This chapter presents three classes of drug therapy, all targeted at the circulatory system. After a brief review of the epidemiology, etiology, and pathophysiology of hypertension, the multiple drug groups used as antihypertensives are described. Drugs used to treat angina pectoris are the second group of drugs described. The third group of agents affecting circulation, antithrombotics, comprises several classes of drugs used to regulate clotting mechanisms.

Hypertension

Epidemiology and etiology

More than 1 billion people worldwide and 1 in every 4 Americans has high blood pressure (≥140/90 mm Hg). Hypertension adversely affects numerous body organs, including the heart, brain, kidney, and eye. Damage to these organ systems resulting from hypertension is termed cardiovascular disease (CVD). Uncontrolled hypertension increases CVD morbidity and mortality by increasing the risk of developing left ventricular hypertrophy, angina, myocardial infarction (MI), heart failure, stroke, peripheral arterial disease, retinopathy, and kidney disease. One of eight deaths can be attributed to hypertension, and the World Health Organization reports that suboptimal blood pressure (systolic blood pressure above 115 mm Hg) is responsible for 62% of cerebrovascular disease and 49% of ischemic heart disease. Blood pressure increases with age, and hypertension is more prevalent in adults older than 65 years. This fact is of great concern because it is estimated that by 2040, 25% of the American population will be older than 65. Hypertension occurs more frequently in men than in women and occurs in more blacks than whites. Evidence suggests that individuals who are normotensive have a greater than 90% lifetime risk for developing hypertension by age 55 (Table 22-1).1

TABLE 22-1

JNC-VII Classification of Blood Pressure for Adults

CATEGORY SYSTOLIC (mm Hg) DIASTOLIC (mm Hg)
Normal <120 <80
Prehypertension 120-139 80-89
Hypertension    
Stage 1 140-159 90-99
Stage 2 ≥160 ≥100

Hypertension is diagnosed by the mean of two or more separate seated blood pressure determinations on different days. The Seventh Report of the Joint National Committee on Detection, Evaluation, and Treatment of High Blood Pressure (JNC-VII), published in 2003, is the most up-to-date guideline compared with other hypertension treatment recommendations.2 The American Heart Association (AHA) released a statement regarding the treatment of hypertension as it relates to the prevention and management of ischemic heart disease, which differed from the JNC-VII recommendations.3 Significant differences between JNC-VII and the AHA statement include expanding the category of the high-risk hypertensive to include patients with known coronary artery disease (CAD) or CAD risk equivalents (e.g., carotid artery disease, peripheral arterial disease, or abdominal aortic aneurysm or a 10-year Framingham risk score of more than 10%).

For high-risk patients, the blood pressure goal remains less than 130/80 mm Hg and less than 140/90 mm Hg for primary prevention. More aggressive target blood pressure of less than 120/80 mm Hg is recommended in patients with left ventricular dysfunction (LVD). First-line treatments for patients with high blood pressure without CAD include four drug classes: thiazide diuretics, angiotensin-converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), and calcium channel blockers (CCBs). β blockers were removed as first-line agents in patients with high blood pressure who do not have CAD. In addition, the AHA does not promote any single agent as preferred first-line therapy if no compelling indications exist.

Individuals with prehypertension, defined by JNC-VII as having a blood pressure reading between 120/80 mm Hg and 139/89 mm Hg, have a 50% greater risk of developing hypertension than individuals with lower blood pressure values. Overall, each increase in systolic blood pressure of 20 mm Hg (or 10 mm Hg in diastolic blood pressure) doubles the risk of a fatal coronary event. Early recognition, lifestyle modifications, and management of prehypertension are essential to decrease the rate of progression to hypertension or prevent hypertension entirely. Most patients require two or more drugs to reach target blood pressure, and when blood pressure is more than 20/10 mm Hg above goal, two drugs should be initiated from the time of diagnosis.

The discussion of hypertension in this chapter is based on the JNC-VII guidelines and 2007 AHA statement. The release of JNC-VIII is not anticipated until Spring 2012, and these guidelines are not reflected in this chapter.

In almost all cases, the etiology of hypertension is unknown, and it is termed either primary hypertension or essential hypertension. The prevalence of secondary hypertension is less than 10%; secondary hypertension includes many disease-induced and drug-induced etiologies. Disease-induced causes of hypertension include Cushing syndrome, hyperparathyroidism, hyperthyroidism, pheochromocytoma, primary aldosteronism, and kidney disease. Drug-induced causes of hypertension include amphetamines, corticosteroids, cyclosporine, erythropoietin, estrogens, nonsteroidal antiinflammatory drugs (NSAIDs) including cyclooxygenase-1 inhibitors (e.g., ibuprofen and naproxen) and cyclooxygenase-2 inhibitors (e.g., celecoxib), pseudoephedrine, sibutramine, tacrolimus, venlafaxine, high sodium–containing over-the-counter (OTC) products (e.g., Alka-Seltzer effervescent antacid tablets), OTC weight loss products (e.g., ephedrine-containing diet pills), and chronic alcohol ingestion.4,5

Pathophysiology

Arterial blood pressure, termed blood pressure, is generated by the interplay between blood flow and the resistance to blood flow. Arterial blood pressure reaches a peak during cardiac systole and a nadir at the end of diastole. Arterial blood pressure is defined hemodynamically as the product of cardiac output (heart rate × stroke volume) and total peripheral resistance. Venous capacitance, which affects the volume of blood (preload), is a major determinant of cardiac output and systolic blood pressure. Arteriolar capacitance (afterload) is a major determinant of total peripheral resistance and diastolic blood pressure. Antihypertensives elicit actions on some or all of the hemodynamic parameters that define arterial blood pressure.

Hypertensive crisis

A patient with blood pressure greater than 180/120 mm Hg is considered to be in a hypertensive crisis. A hypertensive crisis represents either a hypertensive urgency or a hypertensive emergency. Hypertensive urgencies usually signify high blood pressures without signs or symptoms of acute target organ complications; however, patients may present with severe headaches, shortness of breath, nosebleeds, or severe anxiety. In these situations, improvement in blood pressure control can be accomplished over a period of 24 to 48 hours.2 Overaggressive use of intravenous drugs and oral medications can cause too rapid a decrease in blood pressure. Rapid decrease in blood pressure can result in hypoperfusion of organs such as the brain, kidneys, and heart. Oral antihypertensive agents such as captopril, clonidine, and labetalol are routinely used to manage hypertensive urgencies, followed by close observation for several hours. Patients can benefit from antihypertensive medication adjustments if they are found to be noncompliant with taking their medications.

A hypertensive emergency exists when the elevation of blood pressure is accompanied by acute progressing target organ injury. Examples of acute target organ injury include encephalopathy, intracranial hemorrhage, severe retinopathy, renal failure, unstable angina, acute left ventricular failure with pulmonary edema, dissecting aortic aneurysm, and eclampsia. Hypertensive emergencies require admission to an intensive care unit and invasive arterial blood pressure monitoring and immediate, but gradual blood pressure reduction over minutes to several hours with intravenous antihypertensives. The initial goal as outlined in JNC-VII is to reduce the mean arterial pressure (MAP) by no more than 25% within minutes to 1 hour after starting therapy. MAP can be calculated by adding one-third of the systolic blood pressure to two-thirds of diastolic blood pressure:

< ?xml:namespace prefix = "mml" />([⅓ systolic BP] + [⅔ diastolic BP])

image

In the next 2 to 6 hours, the blood pressure must be gradually decreased to 160/100 to 160/110 mm Hg. If the decreased blood pressure is well tolerated by the patient, further reduction of blood pressure toward normal can be attempted over the next 24 to 48 hours. Recommendations differ for patients with ischemic stroke, patients with aortic dissection, and patients awaiting antithrombolytic therapies. Intravenous labetalol and nitroprusside can be used to manage most types of hypertensive emergencies. Depending on other comorbid conditions, alternative intravenous medications can be employed (e.g., nicardipine, esmolol, nitroglycerin, ACEI, and hydralazine). Nitroprusside at high doses or when used for long durations can cause methemoglobinemia. Classic methemoglobin blood is chocolate brown and is without color change despite exposure to air.

Hypertension pharmacotherapy

First-line agents for the treatment of uncomplicated hypertension are thiazide-type diuretics, including ACEIs, ARBs, β blockers, and CCBs.2 Vasodilators, α-blocking agents, α2 agonists, and antiadrenergic agents are considered second-line antihypertensives.2 It is unknown if direct renin inhibitors (DRIs), a new class of antihypertensives, should be considered as first-line agents because long-term morbidity and mortality data are currently unavailable. For stage 1 hypertension, pharmacotherapy should be initiated for most patients with a low dose of a once-daily agent, usually a thiazide-type diuretic, and titrated upward until blood pressure control is achieved or intolerable adverse effects occur. Clinicians should be cognizant that monotherapy achieves effective blood pressure control in only 60% to 70% of patients. Thiazide diuretics profoundly decrease CVD morbidity and mortality, enhance the antihypertensive effects of the other antihypertensives, and are very useful in achieving blood pressure control. For stage 2 hypertension, because a higher response rate may be achieved by initiating low-dose combination antihypertensives, usually a thiazide-type diuretic plus an alternative first-line agent is used. The low-dose combination method may minimize adverse effects and may maximize efficacy and compliance.6,7 Figure 22-1 presents an algorithm for the management of hypertension.2,3

Angiotensin-converting enzyme inhibitors

ACEIs act primarily through suppression of the renin-angiotensin-aldosterone system (RAAS). Because of a lack of renal blood flow, renin is released into the circulation, where it acts on angiotensinogen to produce angiotensin I. In the pulmonary vasculature, angiotensin I is converted by angiotensin-converting enzyme (ACE) to angiotensin II. Angiotensin II is a highly potent endogenous vasoconstrictor that also stimulates aldosterone secretion from the zona glomerulosa cells of the adrenal cortex, contributing to sodium and water retention.8 Angiotensin II also stimulates the release of catecholamines from the adrenergic nerve endings and mediates the release of central sympathetic outflow. ACE is abundant in the endothelial cells of blood vessels and to a lesser extent in the kidneys.

ACEIs block the conversion of angiotensin I to angiotensin II by competing with the physiologic substrate angiotensin I for the active site of ACE (Figure 22-2). The affinity of ACEIs for ACE is approximately 30,000 times greater than for angiotensin I. ACEIs also inhibit kininase which is responsible for the degradation of bradykinin and other vasodilating substances, including prostaglandin E2 (PGE2) and prostacyclin (PGI2), which enhances the antihypertensive effects of these drugs. Because ACEIs are potent antihypertensives in patients with low-renin hypertension, the effects on bradykinin may have an integral role in the mechanism of action of these agents. The hemodynamic effects of ACEIs are a reduction of peripheral arterial resistance, an increase in cardiac output, little or no change in heart rate, an increase in renal blood flow, and unchanged glomerular filtration rate (GFR). ACEIs have mild antihyperlipidemic effects.

Ten ACEIs are available in the United States. ACEIs are the preferred antihypertensives in the setting of systolic dysfunction heart failure, proteinuric kidney disease (diabetic and nondiabetic), and post-MI. ACEIs generally decrease systolic and diastolic blood pressure by 15% to 25%. ACEIs are most effective in normal-renin or high-renin hypertension; however, they are also effective in low-renin hypertension, especially when used at maximal doses. ACEIs are effective alone and in combination with other antihypertensive agents, especially thiazide-type diuretics. The combination of ACEIs and β blockers is less effective in inducing additive blood pressure–lowering effects than other combinations; this is due to the ability of β blockers to reduce renin secretion, which decreases angiotensin II formation.

ACEIs rival diuretics as the most effective first-line antihypertensives to decrease CVD morbidity and mortality in various settings. In contrast to β blockers and thiazide diuretics, ACEIs do not induce glucose intolerance, hyperlipidemia, or hyperuricemia. ACEIs are homogeneous, which means there is very little variability among ACEIs in terms of efficacy and toxicity. With the exception of captopril, all ACEIs are generally administered once or twice daily. Enalaprilat is the only available parenteral ACEI. Table 22-2 presents the pharmacokinetics and dosing guidelines for ACEIs.4,5

TABLE 22-2

Pharmacokinetics and Dosing Guidelines for Angiotensin-Converting Enzyme Inhibitors (ACEIs)

ACEI GENERIC NAME (BRAND NAME) ACTIVE METABOLITE ELIMINATION TOTAL HALF-LIFE OF PARENT DRUG (hr)* DURATION OF ACTION (hr) DOSE RANGE (mg/day) DAILY FREQUENCY EFFECT OF FOOD ON ABSORPTION
Benazepril (Lotensin) Benazeprilat 11%-12% bile 22 24+ 5-80 1 Slightly reduced
Captopril (Capoten) None 95% urine 2 6-10 12.5-450 2-4 Reduced by 30%-40%
Enalapril (Vasotec) Enalaprilat 94% urine and feces 11 24 2.5-40 1-2 None
Enalaprilat (Vasotec IV) None No data 35   1.25-5 Every 6 hours NA
Fosinopril (Monopril) Fosinoprilat 50% urine, 50% feces 12-15 24 10-80 1 Slightly reduced
Lisinopril (Prinivil; Zestril) None 29% urine, 69% feces, 2% unchanged 13 24 10-40 1 None
Moexipril (Univasc) Moexiprilat 13% urine, 53% feces 2-9 24 7.5-30 1-2 Markedly reduced
Quinapril (Accupril) Quinaprilat 60% urine, 37% feces 2-3 24+ 20-80 1-2 Reduced
Perindopril (Aceon) Perindoprilat 96%-78% bile, 4%-12% urine 0.8-1 24 4-16 1 Reduced
Ramipril (Altace) Ramiprilat 60% urine, 40% feces 11-17 24+ 2.5-20 1-2 Slightly reduced
Trandolapril (Mavik) Trandolaprilat 33% urine, 56% feces 24 24+ 1-4 1 Reduced

image

NA, Not applicable.

*Assuming normal renal function.

The most common adverse effect associated with ACEIs is a persistent nonproductive dry cough (20% to 30%). The cough may be due to ACEI-induced accumulation of kinins, prostaglandins, or substance P in the respiratory tract. The cough may develop within days to 1 year after the start of therapy. Antitussives are ineffective in relieving ACEI-induced cough. Cross-reactivity among the ACEIs is absolute; however, ARBs rarely cause cough and may be considered an alternative. ACEI-induced rash is also common; the incidence is 10%, and the reaction is usually transient. The rash often occurs in the upper extremities and is often accompanied by pruritus and erythema. A higher incidence of rash with captopril relative to other ACEIs may be due to the sulfhydryl-containing structure of captopril. All other ACEIs, with the exception of fosinopril (phosphorus-containing), possess a dicarbocyl group. ACEIs are known to cause dysgeusia (6%), manifesting as a metallic or salty taste or loss of taste perception.

ACEIs may cause a slight increase in potassium that is generally inconsequential. The risk of hyperkalemia may be increased, with concomitant use of β blockers, heparin, low-molecular-weight heparin (LMWH), trimethoprim, amiloride, spironolactone, and salt substitutes and in patients with diabetes or renal failure. Orthostatic hypotension is common when initiating ACEI therapy, especially in patients who are in a high-renin state, such as patients who are salt or volume depleted (e.g., patients with heart failure, cirrhosis, or diabetes or receiving diuretics). Patients with bilateral renal artery stenosis, with unilateral stenosis of a solitary functioning kidney, or in a high-renin state (especially patients with heart failure) are susceptible to developing ACEI-induced acute renal failure. Proteinuria, defined as total urinary protein exceeding 1 g/day and, rarely, accompanied by increases in blood urea nitrogen and serum creatinine, may develop in patients receiving high-dose ACEIs or with average ACEI doses and preexisting renal dysfunction. ACEI-induced blood dyscrasias such as neutropenia and agranulocytopenia occur with an incidence of less than 1% and are more common in patients with connective tissue diseases (e.g., systemic lupus erythematosus). ACEIs should be avoided in women of childbearing age because of the potential for fetal and neonatal morbidity and mortality in the second and third trimesters of pregnancy manifesting as skull hypoplasia, hypotension, anuria, and death (pregnancy category D).

Angioedema is rare, occurring in about 1 to 5 of 1000 patients, but it can be life-threatening when accompanied by dyspnea. Angioedema can occur at any time during ACEI therapy, especially when starting and stopping regimens. Angioedema generally manifests in the upper extremities, primarily the face, lips, tongue, glottis, and larynx. ACEI-induced angioedema is an absolute contraindication for the administration of alternative ACEIs and a relative contraindication for ARBs, especially in patients with a history of angioedema with dyspnea or with documented aminopeptidase P deficiency. Angioedema symptoms may be associated with high concentrations of bradykinin. Bradykinin exerts its pharmacologic effects (vasodilation and proinflammation) on bradykinin-2 receptors and is metabolized primarily by ACE, to a lesser extent by aminopeptidase P, and to a minor extent by carboxypeptidase N. Delineating which patients have an aminopeptidase P plasma level deficiency may help predict which patients are predisposed to angioedema.

A significant drug interaction occurs when combining ACEIs with NSAIDs. NSAIDs increase renin release by inhibiting renal vasodilating prostaglandins (PGE2 and PGI2), blunting or negating the antihypertensive effects of ACEIs. NSAIDs less likely to reduce renal prostaglandins and to minimize or circumvent the interaction with ACEIs are sulindac (Clinoril), nabumetone (Relafen), etodolac (Lodine), salsalate (Disalcid), and choline magnesium trisalicylate (Trilisate). ACEIs may increase lithium concentrations and have been associated with life-threatening lithium toxicity. ACEI-induced renal sodium depletion may increase lithium renal tubule reabsorption. Patients receiving this combination should be monitored for symptoms of lithium toxicity such as nausea, vomiting, diarrhea, tremor, and mental status changes. Lithium levels should be monitored before and after initiating the ACEI. A quinapril tablet, in contrast to other ACEIs, contains magnesium carbonate at sufficient concentration to reduce tetracycline absorption 40%. The mechanism of this interaction may be chelation and plausibly may occur with quinolones. To circumvent this interaction, quinapril should be spaced 2 to 6 hours apart from tetracycline and quinolone antimicrobials.

Angiotensin II receptor blockers

Several nonrenin and non-ACE pathways are used for the production of angiotensin II (see Figure 22-2). Nonrenin pathways generate angiotensin II from angiotensinogen via tissue plasminogen activator, cathepsin G, and tonin. Non-ACE enzymes that generate angiotensin II from angiotensin I are cathepsin G, chymostatin-sensitive angiotensin II–generating enzyme, and chymase. ACEIs incompletely block the synthesis of angiotensin II. ARBs are angiotensin II type 1 (AT1) receptor antagonists. AT1 receptors are found in many tissues, such as vascular smooth muscle, myocardial tissue, brain, kidney, liver, uterus, and adrenal glands (cortex and medulla). Many tissues also have an AT2 receptor; however, it is not known to have effects on myocardial hemostasis. ARBs have 1000-fold greater affinity for AT1 receptors than AT2 receptors and generally do not block the AT2 receptor. Because ARBs do not inhibit ACE, they do not interfere with the concentrations of bradykinins and substance P. This kinin-sparing effect may explain why ARBs have a low incidence of inducing cough or angioedema. However, the beneficial effects of kinins, including blood pressure–lowering potency, may be sacrificed.

Seven ARBs are available in the United States. ARBs are indicated for hypertension and can be used to treat heart failure. ARBs have been shown to reduce morbidity, such as target organ damage (e.g., nephropathy) in patients with hypertension, cardiovascular events in patients with systolic heart failure, and progression of nephropathy in patients with type 2 diabetes. In black patients, ARBs and ACEIs may be less potent antihypertensives; however, this can be circumvented by administering maximal doses.

Compared with ACEIs, ARBs are considered as potent or slightly weaker antihypertensive agents. The inhibition of bradykinin by ACEIs may account for its augmented antihypertensive effect. Angiotensin II receptor blockers arguably are considered second-line agents to ACEIs for hypertension and heart failure and are indicated when ACEI-induced cough or other adverse effects are intolerable. However, ARBs may be considered superior to ACEIs in patients with type 2 diabetic nephropathy. ARBs are administered once or twice daily. Using the combination of an ACEI and an ARB has not been well studied; however, its beneficial effects have been observed in patients with heart failure and nephrotic syndrome. Table 22-3 presents the pharmacokinetics and dosing guidelines for ARBs.4,5,8

TABLE 22-3

Pharmacokinetics and Dosing Guidelines for Angiotensin II Receptor Blockers (ARBs)

ARB GENERIC NAME (BRAND NAME) ELIMINATION TERMINAL HALF-LIFE (hr) DOSE RANGE (mg/day) DAILY FREQUENCY EFFECT OF FOOD ON ABSORPTION
Candesartan (Atacand) Ester hydrolysis/O-deethylation 9  8-32 1-2 No effect
Eprosartan (Teveten) 80% unchanged, 20% acyl glucuronide 5-9 400-800 1-2 No effect
Irbesartan (Avapro) CYP2C9, CYP3A4 11-15 150-300 1 No effect
Losartan (Cozaar) CYP2C9, CYP3A4 2 25-100 1-2 Slightly reduced
Olmesartan (Benicar) 35%-50% in urine and remainder in feces 13 20-40 1 No effect
Telmisartan (Micardis) Conjugation to acyl glucuronide 24 20-80 1 Slightly reduced
Valsartan (Diovan) Biliary metabolism 6 80-320 1 Markedly reduced

image

CYP, Cytochrome P450.

The side-effect profile of ARBs seems to be similar to that of ACEIs. ARBs may cause orthostatic hypotension, hyperkalemia, neutropenia, nephrotoxicity, and fetotoxicity. Similar warnings and precautions exhibited with ACEIs should be undertaken for ARBs. ARBs can cause cough; however, the incidence is significantly less than with ACEIs. ARBs cause significantly less angioedema than ACEIs; cross-reactivity has been reported. ARBs are not absolutely contraindicated in ACEI-induced angioedema; however, their use in this setting can be dangerous and should be avoided. Rash and dysgeusia are rarely reported with ARBs.

Losartan is extensively metabolized by the hepatic cytochrome P450 3A4 (CYP3A4) and CYP2C9 isoenzymes to an active carboxylic acid metabolite that is predominantly responsible for the AT1 blockade and antihypertensive effects of losartan. Drugs that induce these enzyme systems (e.g., phenytoin, phenobarbital, carbamazepine, oxcarbazepine, rifampin, and rifabutin) may increase the antihypertensive effects of losartan by increasing the concentration of the active metabolite. Phenobarbital has been shown to decrease the levels of losartan and its metabolite by 20%. Conversely, drugs that inhibit CYP3A4 (e.g., ketoconazole, fluconazole, erythromycin, clarithromycin, fluoxetine, and amiodarone) or CYP2C9 (e.g., amiodarone, cimetidine, and fluoxetine) or CYP3A4 and CYP2C9 simultaneously (e.g., fluoxetine, amiodarone) may decrease the antihypertensive effects of losartan by decreasing the concentration of the active metabolite. However, a study evaluating the effects of cimetidine (CYP3A4 and CYP2C9 inhibitor) on losartan did not yield any changes in the disposition of losartan’s carboxylic acid metabolite.

Telmisartan has been shown to increase digoxin peak plasma concentrations by 50%. Digoxin serum concentrations should be monitored before and after the addition of telmisartan. Several mechanistically similar drug-drug interactions that occur with ACEIs are likely to occur with ARBs, such as with NSAIDs and lithium.

Direct renin inhibitors

DRIs act by inhibiting renin, the enzyme that is the first step of the RAAS (see Figure 22-2). Renin is responsible for the conversion of angiotensinogen to angiotensin I, which is the rate-limiting step in RAAS. Renin inhibition also leads to decreased formation of angiotensin II and aldosterone. However, all agents that inhibit the RAAS, such as ACEIs, have the potential to inhibit feedback inhibition of renin leading to increases in renin and its activity. This effect can be blocked with the use of a renin inhibitor. DRIs can be used alone or in combination with other antihypertensive agents.

Aliskiren (Tekturna) is currently the only DRI available on the market. It is indicated only for the treatment of hypertension. Similar to ACEIs and ARBs, aliskiren is considered a poor antihypertensive agent for black patients. In addition, no studies show that aliskiren is effective in reducing cardiovascular risk. It can be used in combination with any other antihypertensive agents, but it has been studied most comprehensively in combination with ARBs and diuretics.

The most common side effects observed with aliskiren include diarrhea, headache, dizziness, fatigue, upper respiratory tract infection, nasopharyngitis, and back pain. Aliskiren can also cause dry cough, but its incidence is much less than that reported with ACEIs. Similar to other agents that affect the RAAS, aliskiren has been associated with angioedema, and has occurred in patients with and without a history of angioedema with ACEI or ARB therapy. Aliskiren possibly may be fetotoxic and is not recommended for use in pregnant patients (pregnancy category C for the first trimester and pregnancy category D for the second and third trimesters). Rare side effects include increased uric acid levels, renal stones, anemia, rash, renal impairment, myositis, and rhabdomyolysis. Aliskiren monotherapy has a low incidence of hyperkalemia; however, hyperkalemia occurs more frequently when aliskiren is used in combination with ACEIs. It should be used cautiously in combination with other agents that cause hyperkalemia, such as potassium-sparing diuretics and sulfamethoxazole-trimethoprim (Bactrim). Currently, there are no contraindications to use of aliskiren.

Aliskiren is administered once daily at a dose of 150 to 300 mg. It has very poor oral bioavailability; only about 2.5% is absorbed. Absorption of aliskiren is substantially decreased by high fatty meals; patients should always take it the same way: either with or without food. It undergoes minimal hepatic metabolism by CYP3A4. Cyclosporine and itraconazole, which are potent inhibitors of CYP3A4, were shown to increase aliskiren levels significantly and should not be used concomitantly. Other CYP3A4 inhibitors were also shown to increase aliskiren levels, but the clinical significance of their interaction is unknown. Aliskiren has also been shown to reduce the effectiveness of furosemide by 30% to 50%. The effectiveness of furosemide should be monitored when these two agents are used concomitantly. Approximately 25% of the absorbed dose is excreted unchanged in the urine. Most of the unabsorbed drug is excreted in the feces. No dosage adjustments are recommended at this time in patients with renal or hepatic impairment.9,10

Calcium channel blockers

Vascular smooth muscle and cardiac cell contraction depends on free intracellular calcium ion concentration. Calcium enters vascular smooth muscle cells, myocardial cells, and pacemaker cells through voltage-gated L-type and T-type calcium channels. L-channel blockade mediates coronary and peripheral vasodilation and may cause reflex sympathetic activation or a negative inotropic effect. T-channel blockade also mediates coronary and peripheral vasodilation but is devoid of a reflex sympathetic activation. The influx of calcium from extracellular fluid into cells triggers a second messenger, inositol triphosphate, to release stored intracellular calcium from the sarcoplasmic reticulum. This increase in cytosolic calcium results in enhanced binding to the protein calmodulin. A calcium-calmodulin complex activates myosin kinase, promoting the interaction between actin and myosin, culminating in cellular contraction. Conventional calcium channel blockers inhibit only L-channels. The pharmacodynamic effects of the calcium antagonists on smooth muscle, myocardium, or specialized conduction and pacemaker tissues differ among the agents because of different receptor distribution and densities and the drug’s inherent receptor selectivity and affinity.

Nondihydropyridine CCBs include verapamil and diltiazem. Verapamil and, to a lesser extent, diltiazem possess negative chronotropic effects by lowering sinoatrial (SA) node automaticity and decreasing atrioventricular (AV) node conduction; these agents are indicated for the treatment of angina and arrhythmias in addition to hypertension. Verapamil and, to a lesser extent, diltiazem are also potent negative inotropes and may exacerbate heart failure and should be avoided in patients with severe left ventricular dysfunction.

Dihydropyridine CCBs are potent vasodilators; these agents include amlodipine, felodipine, isradipine, nicardipine, nifedipine, and nisoldipine. With the exception of nifedipine, dihydropyridines have negligible chronotropic effects. Immediate-release nifedipine, especially when administered as a liquid (pseudosublingual), causes a potent reflex tachycardia that increases coronary oxygen demand and has been implicated with an increased risk of MI and stroke. Only sustained-release dosage forms of nifedipine are indicated for hypertension.11 Amlodipine and plausibly felodipine may be used in patients with heart failure because these agents do not decrease cardiac contractility. CCBs are very effective antihypertensive agents in both elderly and black patients. Table 22-4 presents the pharmacokinetics and dosing guidelines for calcium antagonists.25

TABLE 22-4

Pharmacokinetics and Dosing Guidelines for Calcium Channel Blockers

CALCIUM ANTAGONIST GENERIC NAME (BRAND NAME) ONSET OF ACTION OF ORAL DOSAGE FORMS (hr) HALF-LIFE (hr) DOSE RANGE (mg/day) DAILY FREQUENCY
Nondihydropyridines
Verapamil (Calan, Isoptin) 0.5 3-7 180-480 3-4
Verapamil SR (Calan SR, Isoptin SR) 0.5 3-7 120-480 1-2
Verapamil ER (Covera-HS) 4-5 2.8-7.4 180-420 Once at bedtime
Verapamil chronotherapeutic oral drug absorption (Verelan PM) 4-5 3-7 100-400 Once at bedtime
Diltiazem (Cardizem) 0.5 3.5 90-360 3-4
Diltiazem ER capsules (Cardizem CD, Cartia XT, Dilacor XR, Diltia XT, Tiazac, Taztia XT) 1 5 90-540 1-2
Diltiazem ER tablets (Cardizem LA) 3-4 6-9 120-540 Once daily (morning or evening)
Dihydropyridines
Amlodipine (Norvasc) 6-12 30-50 2.5-10 1
Felodipine (Plendil) 2-5 11-16 5-20 1
Isradipine (DynaCirc) 2 8 2.5-10 2
Isradipine CR (DynaCirc CR) 2 8 2.5-10 1
Nicardipine (Cardene) 20 minutes 2-4 60-120 3
Nicardipine SR (Cardene SR) 20 minutes 2-4 60-120 2
Nifedipine (Adalat, Procardia)* 20 minutes 2-5 30-120 3-4
Nifedipine LA (Adalat CC, Procardia XL) 20 minutes 7 30-120 1
Nimodipine (Nimotop) ND 1-2 360 Every 4 hours for 21 days
Nisoldipine (Sular) ND 7-12 20-60 1

image

CC, Coat core; CD, controlled delivery; CR, controlled release; ER, extended release; HS, SR, sustained release; LA, long acting; XL, XR, extended release; XT, extended technology.

*Nifedipine (prompt release) is not indicated for hypertension.

Indicated for subarachnoid hemorrhage, not hypertension.

Verapamil (e.g., Covera-HS, Verelan PM) and diltiazem (Cardizem LA) have long-acting formulations that are specifically designed to target the circadian rhythm of blood pressure throughout the day. Many hypertensive patients have a catecholamine surge with a blood pressure peak in the morning between 6 a.m. and 12 p.m., followed by sustained high (but lower than the peak) blood pressures throughout the day and a nadir at night. Most MIs, strokes, dysrhythmias, and venous thromboembolic events occur in the morning hours, in concert with the circadian blood pressure peaks. CCB formulations are generally designed to be dosed at bedtime and begin to release medication in the early morning to achieve a peak effect in the morning hours and a sustained effect during the day.

These novel circadian dosage forms may have limited utility in hypertensive patients who do not have a nadir in blood pressure in the nighttime, or “nondippers.” These formulations leave patients unprotected with a high risk of a coronary event and have not been shown to have better effects on morbidity compared with thiazides and β blockers. Typical hypertensive nondippers (no nighttime nadir) are elderly patients, patients with renal insufficiency, and patients with secondary hypertension. Both verapamil and diltiazem are available in several immediate, extended, and sustained release products. The different dosage formulations of the same drug, with or without circadian effects, are usually not interchangeable and should not be switched on a milligram-to-milligram basis.

The incidence of verapamil-induced, and to a lesser extent diltiazem-induced, constipation is high and often necessitates the use of a stimulant laxative such as bisacodyl or sennosides. Dihydropyridines have potent peripheral vasodilating effects, and they have a high incidence of palpitations, orthostatic hypotension, flushing, headaches, lightheadedness, and syncope. These adverse effects are minimized with long-acting agents. All CCBs may cause peripheral edema, gingival hyperplasia, and gastroesophageal reflux (except diltiazem). CCB-induced peripheral edema does not respond to diuretic therapy and requires discontinuation of the offending agent.

Diltiazem and verapamil inhibit CYP3A4 metabolism and plausibly the P-glycoprotein transport of alfentanil, buspirone, carbamazepine, cyclosporine, digoxin, lovastatin, methylprednisolone, quinidine, simvastatin, and tacrolimus, resulting in higher serum levels and potential toxicity. Verapamil and diltiazem inhibit the hepatic metabolism of theophylline. Although dihydropyridine CCBs are not inhibitors of CYP3A4, they are major substrates of CYP3A4 and may result in significant drug and food interactions through competitive inhibition. Grapefruit juice inhibits the CYP3A4 in the gut and may increase significantly the levels of felodipine, nifedipine, and nisoldipine. Because many CCBs are significantly metabolized by the CYP450 system, CYP450 enzyme inducers, such as carbamazepine, oxcarbazepine, phenobarbital, phenytoin, and rifampin, may lower the serum concentrations of CCBs and compromise efficacy.

β blockers

The antihypertensive effects of β blockers have multiple mechanisms of action and are as follows:

β blockers cannot be used interchangeably with each other. Instead, disease state guidelines dictate which β blockers to use for each comorbidity. β blockers with intrinsic sympathomimetic activity (ISA), including acebutolol, carteolol, penbutolol, and pindolol, cause less reduction of resting heart rate, cardiac output, and peripheral blood flow. ISA may be beneficial in patients with stable angina, bradyarrhythmias, compromised pulmonary function, and peripheral vascular (arterial) disease. Labetalol is an α and β blocker with weak β2 ISA; nevertheless, it is relatively contraindicated in patients with asthma and chronic obstructive pulmonary disease. Labetalol is indicated for hypertension and is often used to manage hypertensive urgencies (oral formulation) and hypertensive emergencies (parenteral formulation). The α and β blocker carvedilol is indicated for patients with hypertension and for patients with mild to moderate heart failure.

The newest agent, nebivolol (Bystolic), is a highly cardioselective third-generation β1 blocker, which also exhibits vasodilatory properties mediated through nitric oxide, resulting in decreased peripheral vascular resistance, increased stroke volume, and preserved cardiac output. It is approximately three times more β1-selective than bisoprolol. It is indicated for the treatment of hypertension and has similar blood pressure reduction effects as atenolol, bisoprolol, ACEIs, ARBs, and CCBs.

β blockers are indicated for hypertension, angina pectoris, cardiac dysrhythmias, secondary prevention of MI, chronic heart failure, and pheochromocytoma. β blockers are no longer considered first-line agents in treatment of essential hypertension but should be reserved as add-on therapy to other antihypertensive agents. β blockers are also used for migraine prophylaxis, hypertrophic subaortic stenosis, tremors, alcohol withdrawal syndrome, prophylaxis of esophageal variceal rebleeding, anxiety, symptoms of thyrotoxicosis, and in combination with α blockers for pheochromocytoma. Table 22-5 presents the pharmacokinetics and dosing guidelines for β blockers.2,3,5,9

TABLE 22-5

Pharmacokinetics and Dosing Guidelines for β Blockers

β BLOCKER GENERIC NAME (BRAND NAME) α BLOCKADE β1 SELECTIVITY ISA LIPID SOLUBILITY HALF-LIFE (hr) DOSE RANGE (mg) DAILY FREQUENCY
Acebutolol (Sectral) 0 + + Low 3-4 200-200 2
Atenolol (Tenormin) 0 + 0 Low 6-9 25-100 1
Betaxolol (Kerlone) 0 + 0 Low 14-24 5-20 1
Bisoprolol (Zebeta) 0 ++ + Low 9-12 25-200 1
Carteolol (Cartrol) 0 0 + Low 6 2.5-10 1
Carvedilol (Coreg) + 0 0 High 7-10 6.25-50 2
Labetalol (Trandate, Normodyne) + 0 0 Moderate 3-5 100-2400 2
Metoprolol (Lopressor) 0 + 0 Moderate 3-5 50-200 1-2
Metoprolol ER (Toprol-XL) 0 + 0 Moderate 3-7 25-200 1
Nadolol (Corgard) 0 0 0 Low 14-24 20-240 1
Nebivolol (Bystolic) 0 +++ ? Low 11-30 5-40 1
Penbutolol (Levatol) 0 0 + High 5 20-80 1
Pindolol (Visken) 0 0 +++ Moderate 3-4 10-60 2
Propranolol (Inderal) 0 0 0 High 4-6 40-240 2
Propranolol LA (Inderal, InnoPran XL) 0 0 0 High 8-10 80-640 1
Timolol (Blocadren) 0 0 0 Low 3-4 20-40 2

image

ER, Extended release; ISA, intrinsic sympathomimetic activity; LA, long acting; XL, extended release; 0, none; +, ++, +++, equals higher degree.

β blockers increase triglycerides and decrease high-density lipoproteins; however, this deleterious effect may diminish after prolonged therapy (1 year). β blockers may cause hyperglycemia and glucose intolerance. These agents can be especially dangerous in diabetics because they mask some of the common symptoms of hypoglycemia, such as palpitations, tremors, and hunger. The use of β blockers in patients with hyperlipidemia or diabetes is acceptable if the lipid and glucose profiles are closely monitored. α and β blockers and agents with ISA are less likely to affect the lipid and glucose profiles adversely.

β blocker–induced pulmonary dysfunction may manifest as bronchospasm, bronchial obstruction, wheezing, dyspnea, cough, and exacerbation of previously stable asthma or chronic airway obstruction. Agents with β1 selectivity, such as atenolol and metoprolol, are less likely to cause pulmonary dysfunction; however, they lose their selectivity with increasing doses. β-blocking agents may exacerbate intermittent claudication and Raynaud phenomenon, and they may cause CNS disturbances, such as vertigo, tiredness, fatigue, somnolence, mental depression, and nightmares. A correlation between the individual lipid solubility of a β blocker and its ability to penetrate the blood-brain barrier and cause CNS adverse effects may exist. Consequently, agents with high lipophilicity, such as propranolol and penbutolol, have a high incidence of CNS adverse effects. β blockers should not be discontinued abruptly because this causes a rebound (pretreatment blood pressure) or overshoot (blood pressure higher than pretreatment) hypertension; the drug should be tapered slowly over 1 to 2 weeks before discontinuing entirely.

Several β blockers, including carvedilol, metoprolol, nebivolol, propranolol, and timolol, are CYP2D6 substrates. Fluoxetine, paroxetine, and sertraline are potent CYP2D6 inhibitors and may significantly increase the effect of the substrate β blocker. Because almost all β blockers are significantly metabolized by the CYP450 system, CYP450 enzyme inducers, such as cigarettes and marijuana, carbamazepine, oxcarbazepine, phenobarbital, phenytoin, and rifampin, may lower serum concentrations of β blockers and compromise efficacy. Atenolol is almost entirely renally eliminated and may be used as an alternative to β blockers that interact via hepatic mechanisms or in patients with liver disease.

Diuretics

Diuretics are divided into the following five classes:

Thiazides and potassium-sparing agents are the only two classes that are used primarily for the management of hypertension. Thiazides, loop diuretics (except ethacrynic acid), and CAIs are sulfonamide-containing agents and may cross-react in patients who have a history of sulfonamide allergy. Sulfonamide-containing agents include sulfonylurea antidiabetics, silver sulfadiazine, tamsulosin, celecoxib, and probenecid. A significant drug-drug interaction occurs when combining diuretics with NSAIDs and combining sodium-depleting diuretics with lithium. The mechanisms of these interactions are similar to the mechanisms of ACEIs and have been discussed previously in this chapter.

Potassium-sparing diuretics

Potassium-sparing agents are weak hypotensive agents when used alone, but they provide an additive hypotensive effect when used in combination with thiazide diuretics. The two agents used clinically are amiloride (Midamor) and triamterene (Dyrenium). These agents are employed primarily for their antikaliuretic effects, to offset the potassium excretion effects of other diuretics. These agents work by blocking sodium channels in the luminal membrane of cells in the distal tubule and collecting duct, attenuating the excretion of potassium, calcium, and magnesium. Both hypokalemia and hypomagnesemia have been implicated as a cause of cardiac arrhythmias; there is an advantage to adding these agents to diuretic antihypertensive therapy. The magnesium-sparing effects of potassium-sparing diuretics may be an added benefit compared with a diuretic plus a potassium supplement.

Both potassium-sparing diuretics can cause gastrointestinal side effects, such as dyspepsia, abdominal cramps, nausea, and diarrhea; CNS side effects, such as mental confusion, lethargy, headache, and dizziness; and hematologic, dermatologic, and musculoskeletal (leg cramps) adverse effects. Triamterene has been associated with interstitial nephritis and nephrolithiases; the incidence may be 1 in 200. Triamterene is photosensitizing, which may be additive when combined with phototoxic sulfonamide-containing thiazide diuretics. Triamterene may cause hyperuricemia and hyperglycemia.

Thiazide and thiazide-like diuretics

Thiazide diuretics increase sodium and chloride excretion by interfering with their reabsorption in the distal tubule; a mild diuresis of slightly concentrated urine results. Excretion of potassium, bicarbonate, magnesium, phosphate, and iodide is also increased, whereas calcium excretion is decreased. Although thiazides decrease extracellular fluid volume, antihypertensive activity is caused primarily by direct vasodilation. Thiazides are indicated for hypertension, chronic edema, chronic heart failure, and ascites. Thiazides generally take 2 to 4 weeks to elicit their full pharmacologic effect. Thiazides have a dose-ceiling effect, at which point the antihypertensive effects do not increase despite dosing increases; however, the toxic effects do not have a dose-ceiling effect.

Because thiazides cause hypercalcemia, they may be a useful adjunct in the management and prevention of osteoporosis. Although chlorthalidone, indapamide, and metolazone do not possess the benzothiadiazine structure, pharmacologically they act like thiazide diuretics—they are thiazide-like in structure and activity. Thiazide diuretics lose their antihypertensive potency in patients with a creatinine clearance less than 30 mL/min. Indapamide retains its potency, however, in patients with a creatinine clearance greater than 15 mL/min. Metolazone is the only thiazide-like diuretic that retains potency in patients with a creatinine clearance less than 15 mL/min. Despite the thiazide-like structure of metolazone, its pharmacologic effects are similar to those of loop diuretics. Metolazone is often added to a loop diuretic in patients with diuretic resistance, achieving a synergistic diuretic effect. Mykrox tablets are a formulation of metolazone with a higher bioavailability than conventional metolazone, resulting in a more rapid diuretic effect; Mykrox is not therapeutically equivalent to Zaroxolyn. Table 22-6 presents the pharmacokinetics and dosing guidelines for thiazide and thiazidelike diuretics.2,4,5

TABLE 22-6

Pharmacokinetics and Dosing Guidelines for Thiazides and Thiazide-Like Diuretics

THIAZIDE/THIAZIDE-LIKE DIURETIC GENERIC NAME (BRAND NAME) BIOAVAILABILITY PEAK EFFECT (hr) DURATION OF DIURESIS (hr) HALF-LIFE (hr) DOSE RANGE (mg/day) DAILY FREQUENCY
Chlorothiazide (Diuril) 10-20 2 (PO), 0.5 (IV) 6-12 (PO), 2 (IV) 1-2 500-2000 1-2
Chlorthalidone (Hygroton) 65 2 24-72 35-55 15-200 1
Hydrochlorothiazide (Esidrix, HydroDIURIL, Oretic, Microzide) 65-75 4-6 6-12 2.5-4.5 25-100 1-3
Indapamide (Lozol) 95 2 24-36 14-18 1.25-5 1
Metolazone (Zaroxolyn) 65 2 12-24 6-20 5-20 1
Metolazone (Mykrox)   2-4 12-24 14 0.5-1 mg 1

image

IV, Administered intravenously; PO, administered orally.

Common side effects observed with thiazide and thiazide-like diuretics include hypokalemia, hypomagnesemia, hypercalcemia, hyperuricemia, hyperglycemia, hyperlipidemia, and sexual dysfunction. These abnormalities are dose-related and may be minimized by using low-dose agents such as chlorthalidone, 12.5 to 25 mg daily, or hydrochlorothiazide, 12.5 mg twice daily. Less common thiazide-induced adverse effects include dyspepsia, rashes, photosensitivity, thrombocytopenia, and pancreatitis.

Loop diuretics

Loop diuretics, often referred to as high-ceiling diuretics, act principally at the thick ascending limb of the loop of Henle, where they decrease sodium reabsorption by competing for the chloride site on the Na+-K+-2Cl symporter (a transport molecule). Excretion of sodium, chloride, potassium, hydrogen ion, calcium, magnesium, ammonium, bicarbonate, and possibly phosphate is enhanced. Diuretics such as thiazides have a limited diuretic potency with a plateau effect because they act primarily at sites past the ascending limb; only a small percentage of the filtered load reaches these more distal sites. Because more than 25% of the filtered load is reabsorbed in the ascending limb, loop diuretics are highly efficacious with increasing doses, and this is why they are termed high-ceiling diuretics.

Loop diuretics are indicated for chronic heart failure, ascites with or without hepatic cirrhosis, renal failure, pulmonary edema, hypercalcemia, hypermagnesemia, and syndrome of inappropriate antidiuretic hormone. Loop diuretics are second-line diuretics in the management of hypertension; however, they are superior to thiazide diuretics in diuresis and decreasing blood pressure for patients with renal insufficiency. Table 22-7 presents the pharmacokinetics and dosing guidelines for oral loop diuretics.2,4,5

TABLE 22-7

Pharmacokinetics and Dosing Guidelines for Oral Loop Diuretics

LOOP DIURETIC GENERIC NAME (BRAND NAME) BIOAVAILABILITY (%) ONSET (hr) DURATION (hr) HALF-LIFE (hr) DOSE RANGE (mg/day) DAILY FREQUENCY
Bumetanide (Bumex) 70-95 0.5-1 5-6 0.8 ± 0.2 0.5-10 1
Ethacrynic acid (Edecrin) 100 0.5 6-8 2-4 50-200 1-2
Furosemide (Lasix) 60 0.5-1 6-8 0.5-1.1 40-240 1-2
Torsemide (Demadex) 80 0.5-1 1 2-4 5-200 1

image

Loop diuretics are very potent and consequently may cause severe dehydration, hypotension, hypochloremic alkalosis, and hypokalemia. Loop diuretics should not be administered at bedtime because the patient will have to urinate frequently, causing sleep disturbances. Loop diuretics may cause hyperglycemia (not reported with bumetanide), hyperuricemia, dyspepsia, photosensitivity, and ototoxicity. Ethacrynic acid is the most auditory ototoxic loop diuretic and should be considered only for patients refractory to other loop diuretics or when there is a history of a life-threatening sulfonamide allergy.

Aldosterone antagonists

Spironolactone (Aldactone) and eplerenone (Inspra) are aldosterone antagonists that exert their effect on the late distal tubule and collecting duct. Spironolactone, a weak diuretic, is used primarily for its aldosterone antagonist effects. Spironolactone is indicated for hypertension, management of hepatic cirrhosis (diuretic of choice), primary hyperaldosteronism, hypokalemia, and heart failure. For hypertension, spironolactone is used in combination with other antihypertensives or to spare potassium when administered with diuretics. The chemical structure of spironolactone resembles the structure of the corticosteroids and may explain its sexual adverse effects, such as impotence, decreased libido, gynecomastia, deepening of the voice, menstrual irregularities, and hirsutism. Other spironolactone-induced adverse effects include diarrhea, gastritis, skin rashes, drowsiness, lethargy, ataxia, headaches, and confusion. Similar to the other potassium-sparing diuretics, spironolactone may cause hyperkalemia. Table 22-8 presents the pharmacokinetics and dosing guidelines for the aldosterone antagonists.2,4,5

TABLE 22-8

Pharmacokinetics and Dosing Guidelines for Aldosterone Antagonists

ALDOSTERONE ANTAGONIST GENERIC NAME (BRAND NAME) ACTIVE METABOLITE ELIMINATION TOTAL ONSET OF ACTION (hr) PEAK RESPONSE DURATION OF ACTION (hr) HALF-LIFE OF PARENT DRUG (hr) DOSE RANGE (mg/day) DAILY FREQUENCY EFFECT OF FOOD ON ABSORPTION
Spironolactone (Aldactone) Canrenone 47%-57% renal, 35%-41% fecal 2-4 6-8 hours 16-24 1.4 25-400 1-2 Increased
Eplerenone (Inspra) None 67% renal, 32% fecal 1-2 4 weeks 24 3.5-6 50-100 1-2 No effect

image

Eplerenone is indicated for heart failure after MI and hypertension. Similar to spironolactone, eplerenone blocks the mineralocorticoid receptor, but, in contrast to spironolactone, it does not block the progesterone or androgen receptor, minimizing the sexual adverse effects such as gynecomastia, breast pain, impotence, and menstrual irregularities. Eplerenone has a higher incidence of severe hyperkalemia, especially in patients with reduced renal function. Because of the risk of severe hyperkalemia, eplerenone is contraindicated in all patients with potassium values greater than 5.5 mEq/L or creatinine clearance less than 30 mL/min and in hypertensive patients with type 2 diabetes and microalbuminemia, concomitant use of potassium supplements or potassium-sparing diuretics, or serum creatinine greater than 2 mg/dL in men and greater than 1.8 mg/dL in women or a creatinine clearance less than 50 mL/min. Vigilant monitoring of serum potassium levels is necessary when eplerenone is administered with ACEIs, ARBs, or β blockers. Eplerenone is a CYP3A4 substrate; CYP3A4 inhibitors such as verapamil, diltiazem, erythromycin, fluconazole, and saquinavir may increase eplerenone levels by 50%. Grapefruit juice may also increase levels of eplerenone but to a lesser extent (approximately 25%). It is recommended to initiate eplerenone at a lower dose of 25 mg/day in patients who are taking CYP3A4 inhibitors.

Centrally acting adrenergic agents

The centrally acting adrenergic agents, or α2 agonists, decrease blood pressure by affecting cardiac output and peripheral resistance; they are negative inotropes and chronotropes. α2 agonists stimulate brainstem α2 receptors, resulting in a decrease in sympathetic outflow from the CNS. α2 agonists are very effective antihypertensives; however, they are not considered first-line therapy because of their side-effect profile. They have a high incidence of anticholinergic-like side effects, such as sedation, blurred vision, dry mouth, constipation, and urinary retention, and CNS side effects, such as drowsiness, fatigue, headaches, depression, psychosis, and nightmares. Long-term use of these agents results in sodium and fluid retention and almost always necessitates the use of concomitant diuretics; this is especially seen with methyldopa. α2 agonists are not recommended for noncompliant patients and should never be withdrawn abruptly because of the risk of either rebound hypertension or overshoot hypertension.

The most effective and least toxic α2 agonist is the clonidine transdermal therapeutic system (Catapres-TTS), which achieves sustained levels of clonidine for 7 days. The sustained clonidine levels avoid the peaks and troughs associated with the prompt release dosage form, and treatment is relatively devoid of the troublesome anticholinergic and CNS side effects. The clonidine patch is applied to a hairless area of intact skin on the upper torso. On the initial application, the clonidine patch takes 2 to 3 days to achieve target blood levels and a therapeutic effect. It is recommended to coadminister clonidine oral tablets along with the patch for the first 2 to 3 days of therapy. The most common adverse effects of the patch are local skin rashes and irritation. Table 22-9 presents the pharmacokinetics and dosing guidelines for α2 agonists.2,4,5

TABLE 22-9

Pharmacokinetics and Dosing Guidelines for Centrally Acting Adrenergic Agents (α2 Agonists)

α2 AGONIST GENERIC NAME (BRAND NAME) ONSET OF ACTION (hr) PEAK EFFECT (hr) DURATION OF ACTION (hr) HALF-LIFE (hr) ELIMINATION DOSE RANGE (mg/day) DAILY FREQUENCY
Methyldopa (Aldomet) 4-6 6-9 24-48 1.25 Renal (biphasic) 500-2000 2-3
Clonidine (Catapres) 0.5-1 3-5 24 6-20 Renal (40%-60%) 0.1-2.4 2-4
Guanfacine (Tenex) 2.5 6 24 17 Renal (50%) 1-3 Once at bedtime
Guanabenz (Wytensin) 1 2-5 6-8 7-10 Renal (70%-80%) 4-32 2

image

α1-adrenergic antagonists

α1-adrenergic receptor antagonists selectively block postsynaptic α1 receptors. Total peripheral resistance is reduced through arterial and venous dilation; these agents decrease both preload and afterload and cause a potent first-dose sympathetic reflex increase in heart rate and renin activity.12 α1-adrenergic antagonists cause a first-dose phenomenon that manifests with orthostatic hypotension, tachycardia, palpitations, dizziness, headaches, and syncope. After several doses, despite persistent vasodilation, tolerance to the first-dose phenomenon develops, and heart rate, renin, and cardiac output return to normal. To minimize the first-dose phenomenon, initial doses of α1-adrenergic antagonists should be low and administered at bedtime.

α1-adrenergic antagonists are indicated for hypertension, benign prostatic hyperplasia (BPH), heart failure, and Raynaud vasospasm except for uroselective α1-adrenergic antagonists (tamsulosin and alfuzosin), which are indicated only for BPH. In contrast to other antihypertensives, α1-adrenergic antagonists have favorable effects on the lipoprotein profile and may decrease triglycerides and low-density lipoproteins and increase high-density lipoproteins by 5% to 10%, making them an ideal drug of choice. However, the ALLHAT (Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial) study compared doxazosin with other antihypertensives (chlorthalidone) and revealed a 25% higher incidence of combined cardiovascular morbidity in patients receiving doxazosin.13 A higher incidence of doxazosin-induced stroke, heart failure, angina, and coronary revascularization was reported. On the basis of the results of this study, α1-adrenergic antagonists are considered second-line antihypertensive therapy. Table 22-10 presents the pharmacokinetics and dosing guidelines for α1-adrenergic antagonists.2,4,5

TABLE 22-10

Pharmacokinetics and Dosing Guidelines for α1-Adrenergic Receptor Antagonists

α1 ANTAGONIST GENERIC NAME (BRAND NAME) ELIMINATION ROUTES PEAK (hr) DURATION (hr) HALF-LIFE (hr) DOSE RANGE (mg/day) DAILY FREQUENCY
Doxazosin (Cardura) 63% feces, 9% urine 6 18-36 11 1 to 16 1
Prazosin (Minipress) 90% feces, 10% urine 1.5 8-10 2 3-40 2-3
Terazosin (Hytrin) 60% feces, 40% urine 2 24 14 120 1-2

image

Antiadrenergic agents

Reserpine, guanethidine (Ismelin), and guanadrel (Hylorel) are antiadrenergic antihypertensive agents. All three of these agents are second-line antihypertensives. Reserpine works by binding to storage vesicles of peripheral and central postganglionic adrenergic neurons and depleting norepinephrine. Subsequently, reserpine renders the neuronal storage vesicles dysfunctional. Reserpine may cause sedation, depression, suicidal ideation, psychosis, peptic ulcer disease, and nasal stuffiness. The side effects of reserpine can be minimized with low yet effective antihypertensive doses (0.25 mg or less). Guanethidine and guanadrel are postganglionic sympathetic inhibitors that produce a selective block of efferent peripheral sympathetic pathways. Guanethidine and guanadrel act as substitute neurotransmitters by replacing norepinephrine in the neuronal storage vesicle. Guanethidine and guanadrel cause similar adverse effects, such as orthostatic hypotension, sexual dysfunction, and diarrhea that can be occasionally explosive. The antihypertensive effects of these agents may be diminished when combined with tricyclic antidepressants, amphetamines, and ephedrine.

Vasodilators

The two common vasodilators used in the management of hypertension are hydralazine (Apresoline) and minoxidil (Loniten). Because of their side-effect profile, the vasodilators are second-line antihypertensive agents. Hydralazine is also indicated for heart failure and has been used for angina. These agents reduce total peripheral resistance by a direct action on vascular smooth muscle, increasing intracellular concentrations of cyclic guanosine 3′,5′-monophosphate (cGMP). These vasodilators are so potent that they cause a profound activation of baroreceptors, leading to reflex tachycardia, renin release, and an increase in cardiac output. To minimize tachycardia and fluid retention, these agents are often administered concomitantly with a β blocker and a loop diuretic, respectively. Hydralazine has been associated with peripheral neuropathy and drug-induced systemic lupus erythematosus–like syndrome. When hydralazine is administered with food, its bioavailability may double and may cause cardiac toxicity. Hydralazine should be administered consistently with or without food. Minoxidil-induced adverse effects include hirsutism, nausea and vomiting, and pericardial effusions.

Angina

Epidemiology, etiology, and pathophysiology

Ischemic heart disease can manifest as many clinical variants such as stable exertional angina; unstable (rest, preinfarction, crescendo) angina; coronary vasomotion; vasospasm associated with atypical, variant, or Prinzmetal angina; silent myocardial ischemia; or MI. Angina pectoris (chest pain) is a symptom or marker of myocardial ischemia. Ischemia is defined as a lack of oxygen and decreased or no blood flow to the myocardium. From 2002-2006, greater than 10 million Americans older than 20 years were diagnosed with angina pectoris.14 Women often initially present with angina, whereas men present with MI. CAD, when present, tends to be less severe in women than men.

Angina pectoris can manifest with a heavy weight or pressure on the chest, a burning sensation, or shortness of breath. The chest tightness or pressure can occur over the sternum, left shoulder, and lower jaw. Chest pain can be precipitated by physical exercise, cold environment, or emotional stress (anger). The duration of pain intensity may range from a few minutes to half an hour. During angina, an imbalance of myocardial oxygen supply and myocardial oxygen demand occurs. Factors that increase myocardial oxygen demand include increased heart rate, increased systolic wall force or tension, or increased contractility. Factors that decrease myocardial oxygen supply include a decrease in the concentration of oxygen (e.g., anemia), a decrease in coronary blood flow (e.g., thrombus), or inability of the myocardium to extract oxygen from the blood.

Pharmacotherapy

Pharmacotherapy for angina pectoris includes nitrates, β blockers, CCBs, and ranolazine (Ranexa). Ranolazine was approved by the U.S. Food and Drug Administration (FDA) in 2006 for the treatment of chronic stable angina in combination with amlodipine, β blockers, or nitrates.15 For the management of vasospastic and chronic stable angina, diltiazem, verapamil, amlodipine, and nifedipine are indicated. For the management of angina, β blockers are usually dosed to achieve a resting heart rate of 50 to 60 beats/min and a maximal exercise heart rate of 100 beats/min. Goal blood pressure for patients with documented CAD (i.e., chronic stable angina, unstable angina, non–ST segment elevation MI, ST segment elevation MI) is less than 130/80 mm Hg because of increased risk for mortality. All patients with angina should receive daily aspirin (75 to 100 mg/day) to prevent MI.1619

Nitrates

Nitroglycerin reduces myocardial oxygen demand by causing venodilation of coronary arteries and collaterals, resulting in decreased end-diastolic pressures. Venous effects predominate; however, nitroglycerin can affect arteries at high doses. The cellular mechanism of action of nitrates is depicted in Figure 22-3. Nitrates are indicated for acute treatment or prophylaxis of angina, acute MI, acute heart failure, low-output syndromes, and hypertension (intravenous). Nitrates may be administered by various routes and are readily available in multiple preparations, including oral, intravenous, ointment, transdermal, translingual, and sublingual tablets. Sublingual nitroglycerin is indicated for acute anginal relief. Sublingual nitroglycerin has an onset of action of minutes and duration of action of 30 minutes. Sublingual nitroglycerin should be administered every 5 minutes until relief is obtained. If pain relief is not achieved after three doses in 15 minutes, emergency care should be sought. This algorithm is recommended for patients who were previously prescribed nitroglycerin. For patients who were never prescribed nitroglycerin, it is recommended that patients should seek emergency care if symptoms worsen or persist 5 minutes after taking the first sublingual nitroglycerin.19 Sublingual tablets must always be stored in their original glass container, and any unused tablets should be discarded 6 months after the original container is opened because of loss of potency. Other forms of nitroglycerin are isosorbide dinitrate (Isordil) and isosorbide mononitrate (Imdur, Ismo, and Monoket). Table 22-11 presents the pharmacokinetics and dosing guidelines of nitrates.

Angina pectoris

Chronic heart failure

Perioperative hypertension

Isosorbide dinitrate Isosorbide mononitrate 45-60 6-12

image

ER, Extended release; IV, intravenous; PVC, polyvinyl chloride.

Serious adverse reactions to nitrates are uncommon and involve mainly the cardiovascular system. The most frequent adverse effects include tachycardia, palpitations, postural hypotension, dizziness, flushing, and headache. Case reports of clinically significant methemoglobinemia are rare at conventional doses. Methemoglobinemia formation is dose-related and occurs by the nitrite ion reacting with the ferrous hemoglobin. Tolerance to the vascular and antianginal effects may occur after 24 hours of continuous therapy with any formulation. Because most evidence supports the central role of cGMP stimulation in nitrate-induced vasodilation, it has been suggested that the tolerance results from sulfhydryl depletion at the nitrate receptor. Sulfhydryl depletion leads to reduced S-nitrosothiol production and a decreased production of cGMP. Theoretically, administration of a sulfhydryl donor, such as N-acetyl cysteine or captopril, may restore vascular response to nitrates. Increasing doses of nitroglycerin overcome tolerance, but this is short-lived. To circumvent nitrate tolerance, a nitrate-free interval of 10 to 14 hours is suggested. Nitrates are contraindicated in patients concomitantly taking phosphodiesterase type 5 inhibitors for erectile dysfunction, such as sildenafil (Viagra), vardenafil (Levitra), and tadalafil (Cialis), because of pronounced potentiation of nitric oxide resulting in profound hypotension, MI, and even death.

Ranolazine

Ranolazine is indicated for the treatment of patients with chronic angina who have not achieved an adequate response with other antianginal drugs. In 2007, the American College of Cardiology Foundation (ACCF) and the American Heart Association (AHA) suggested that ranolazine may be safely administered for symptomatic relief after unstable angina or non–ST segment elevation MI, but it does not seem to reduce significantly cardiovascular death, MI, or recurrent ischemia. Ranolazine provides antiischemic effects that complement the benefits of CCBs, β blockers, and nitrates. Although the exact mechanism of how ranolazine exerts its antianginal and antiischemic effects is unknown, it is speculated that it selectively inhibits the late phase of the inward sodium channel in ischemic myocytes resulting in decreased myocardial oxygen consumption.20 Ranolazine increases exercise tolerance, which reduces angina frequency and the need for emergent nitroglycerin interventions. In contrast to standard antianginal medications, ranolazine does not alter blood pressure or heart rate. The initial adult dose of ranolazine extended release tablets is 500 mg twice daily, with a maximal dose of 1 g twice daily.

Adverse reactions observed with ranolazine include dizziness, palpitations, headache, constipation, nausea, abdominal pain, and peripheral edema. Small, reversible increases in serum creatinine and blood urea nitrogen (BUN) have also been observed without the incidence of renal toxicity. Ranolazine is excreted primarily in the urine (75%) and to a lesser extent in the feces (25%); however, the manufacturer suggests that no dosage adjustments are needed in patients with renal impairment. Nevertheless, patients with renal impairment taking ranolazine were observed to have a 15-mm Hg increase in blood pressure—frequent blood pressure monitoring is prudent in such patients. Ranolazine can prolong the cardiac Q–Tc interval (Q–T interval [duration of ventricular electrical activity], corrected for heart rate) and place patients at risk of torsades de pointes; this effect is dose-dependent. A dose of 1 g twice daily prolongs the Q–Tc by 6 msec and is more pronounced with hepatic dysfunction. Ranolazine is contraindicated in patients with any degree of hepatic dysfunction or who are receiving other agents that prolong the Q–Tc. Baseline and follow-up electrocardiography (ECG) should be completed during ranolazine therapy.

Ranolazine is extensively metabolized in the gut and liver by CYP3A4 and to a lesser extent by CYP2D6. CYP3A4 inhibitors, such as ketoconazole, fluconazole, macrolides, diltiazem, and verapamil, can increase significantly the plasma levels of ranolazine and are contraindicated. Ranolazine is a substrate of P-glycoprotein, and it should not be taken with verapamil, a known inhibitor of P-glycoprotein. Ranolazine is a P-glycoprotein inhibitor and has been shown to increase the plasma concentration of digoxin by 1.5-fold. Ranolazine is also an inhibitor of CYP3A4 and CYP2D6, plausibly increasing the plasma levels of drugs that are substrates of these enzymes, such as statins, tricyclic antidepressants, and antipsychotics.

Antithrombotic agents

Antithrombotics may be defined as agents that prevent or break up blood clots in conditions such as thrombosis or embolism. Three categories of antithrombotic agents are currently available in the United States: anticoagulants, antiplatelets, and thrombolytics. Anticoagulant agents work by preventing the formation of the fibrin clot and preventing further clot formation in already existing thrombi. Antiplatelet agents inhibit the action of platelets in the initial stage of the clotting process. Thrombolytics break up thrombi by degrading fibrin. Box 22-1 lists currently available antithrombotic agents.21

Formation and elimination of acute coronary thrombus

Under normal conditions, the body maintains an equilibrium state between clot formation (thrombosis) and clot breakdown (fibrinolysis).22 Thromboses are initiated by an injury to the endothelial wall of a coronary vessel. When injury occurs, the anticoagulated endothelial surface is disrupted, and the highly procoagulant subendothelial surface is exposed. Instantaneously, platelets aggregate in response to the release of chemotactic substances, such as thromboxane A2, followed by platelet adhesion to the subendothelial vessel surface, representing the initial step in clot formation. Platelet adhesion is mediated mainly by von Willebrand factor. von Willebrand factor is present in the subendothelium and is actively recruited when the subendothelium is injured. Adhered platelets are exposed to many subendothelial proteins, such as collagen and thrombin. Collagen and thrombin also promote platelet activation. Activated platelets release platelet agonists such as adenosine diphosphate (ADP), norepinephrine, serotonin, and arachidonic metabolites, mitigating and amplifying platelet aggregation and forming an unstable thrombus or platelet plug.

The most important consequence of platelet activation is the expression of platelet receptor glycoprotein (GP) IIb/IIIa on the platelet’s surface, allowing binding to fibrinogen. Fibrinogen binds to the two GP IIb/IIIa molecules, causing a cross-linking of receptors on adjacent platelets and initiating platelet aggregation. Triggers affecting platelet aggregation and their antagonists are depicted in Figure 22-4. Fibrinogen is converted into fibrin monomers by the action of thrombin; this is the final step in clot formation. Homeostasis is complete when the fibrin clot becomes insoluble within the vessel. This stable fibrin clot is the end result of the coagulation cascade. Under normal conditions, multiple inhibitors and control mechanisms keep these reactions localized to the site of the injury.

The fibrin clot ultimately must be removed for hemostasis to be maintained. Activation of the fibrinolytic system by tissue plasminogen activators (t-PAs), which are present in most body fluids and tissues, results in the conversion of plasminogen to plasmin, initiating the dissolution of fibrin and fibrinogen. The breakdown of fibrinogen and fibrin results in polypeptides termed fibrin split or fibrinogen degradation products (FDPs). FDPs are anticoagulant substances that can cause bleeding if fibrinolysis becomes uncontrolled and excessive. D-dimers are fragments of plasmin-digested, cross-linked fibrin that increase in concentration after the onset of fibrinolysis. Blood testing for D-dimer fragments may assist in the diagnosis of pathogenic venous thromboembolism. The extrinsic and intrinsic pathways of the coagulation system are depicted and described in Figure 22-5.2,3,18

Anticoagulant agents

Heparins: unfractionated heparin and low-molecular-weight heparin

Heparin is a nonionic sulfated glycosaminoglycan anticoagulant naturally present in the secretory granules of human mast cells. When heparin is released from mast cells, it is ingested and destroyed by macrophages. Heparin is not detectable in plasma except in pathologic circumstances (e.g., mastocytosis). Commercially available unfractionated heparin (UFH), or simply heparin, is indicated for prevention and treatment of venous thromboembolism, prevention and treatment of pulmonary embolism, treatment of atrial fibrillation with embolization, diagnosis and treatment of disseminated intravascular coagulation, and prophylaxis and treatment of peripheral arterial embolism.

Heparin is extracted from porcine intestinal mucosa or bovine lungs; however, because of the high propensity of thrombocytopenia with the bovine lung derivative, only the porcine derivative is routinely employed in practice. Heparin serves as a catalyst that accelerates the rate of the thrombin-to-antithrombin III reaction by at least 1000-fold by serving as a catalytic template to which both bind, resulting in a ternary complex (heparin, thrombin, and antithrombin). Antithrombin III is a large protein (58,000 Da) that is synthesized in the liver and is known as a suicide substrate. Heparin is a high-molecular-weight complex mucopolysaccharide containing specific pentasaccharide units and approximately 45 monosaccharide side chains with a mean molecular mass of 12,000 Da (range 5000 to 30,000 Da).23

Most of the monosaccharide side chains of UFH are more than 18 monosaccharides long and are necessary to form the ternary complex. Heparin molecules that possess less than 18 monosaccharide units (less than 5400 Da) do not catalyze the thrombin-to-antithrombin III reaction. However, the heparin molecules that include less than 18 monosaccharides catalyze a conformational change on antithrombin III that inhibits the effects of factor Xa (Stuart factor) and does not require a ternary complex. LMWHs are generally about 4500 Da (range 1000 to 10,000 Da) and contain 15 monosaccharide units and do not form a ternary complex. Their anticoagulant activity is exhibited via factor Xa inhibition. Because factor Xa occurs earlier in the coagulation cascade, the inhibition of a single molecule of factor Xa prevents thousands of thrombin molecules from forming. The anti–factor Xa/anti–factor IIa ratio of UFH is 1:1; the anti–factor Xa/anti–factor IIa ratio of LMWH ranges from 2:1 (dalteparin) to 3.8:1 (enoxaparin). Heparin also inhibits the conversion of fibrinogen to fibrin and inhibits the activation of factor XIII, preventing the formation of a stable fibrin clot. LMWH is postulated to suppress von Willebrand factor, which increases platelet aggregation, and to stimulate the release of tissue factor pathway inhibitor, which inhibits factor Xa.

LMWHs include dalteparin (Fragmin), enoxaparin (Lovenox), and tinzaparin (Innohep). In contrast to LMWHs, UFH binds extensively to plasma proteins such as glycoproteins, vitronectin, lipoproteins, fibrinogen, platelet proteins such as platelet factor-4, acute-phase reactant proteins, and endothelial cells, yielding poor UFH bioavailability and an unpredictable effect. The predictable bioavailability of LMWHs allows for subcutaneous administration for all indications. UFH is administered subcutaneously for venous thromboembolism prophylaxis but must be administered as a continuous infusion for serious indications such as MI and to minimize the risk of hemorrhage.

High-molecular-weight heparin (UFH) is cleared faster and requires more frequent dosing or an intravenous continuous infusion. The half-life of UFH is approximately 30 to 60 minutes, whereas the half-life of LMWH is 4 to 5 hours, allowing for once-daily or twice-daily LMWH administration. The onset of action of heparin is within 6 hours of initiation of a continuous infusion. LMWH time to peak anti–factor Xa activity is 2 to 5 hours. Each commercially available LMWH is synthesized by different mechanisms, possesses moderately different pharmacokinetic and pharmacodynamic characteristics, and has different FDA-approved indications and dosing regimens; these agents are not interchangeable. Table 22-12 presents the pharmacokinetic properties and dosing parameters for all heparins.

Enoxaparin (Lovenox) 3500-5500 3.8:1 129-180 Fondaparinux (Arixtra) 1728 NA   Tinzaparin (Innohep) 5500-7500 2.8:1 180-240 No labeled dose Unfractionated Heparin (UFH) 10,000-15,000 1:1 30-150* 5000 U every 8 hours

image

aPTT, Activated partial thromboplastin time; CrCl, creatinine clearance; DVT, deep vein thrombosis; IV, intravenous administration; MI, myocardial infarction; NA, not available; NSTEMI, non–ST segment elevation myocardial infarction; PE, pulmonary embolism; SQ, subcutaneous administration; STEMI, ST segment elevation myocardial infarction; THR, total hip replacement; TKR, total knee replacement; VTE, venous thromboembolism

*Half-life of UFH has saturable binding, and half-life increases with doses more than 400 U/kg.

Activated partial thromboplastin time (aPTT) is used to monitor the effects of heparin because it is sensitive to the inhibitory effects of thrombin, factor Xa, and factor IXa and correlates with heparin levels. When the concentration of plasma heparin is 0.1 to 1 U/mL, aPTT and thrombin time are prolonged. The goal of heparin therapy is to prevent unwanted clotting without an increased risk of hemorrhage. This goal may be accomplished by maintaining aPTT between 2 and 2.5 times the upper limit of the control value. aPTT should not be used to monitor LMWHs. The effect of LMWHs may be monitored on the basis of anti–factor Xa levels; however, because the relationship between anti–factor Xa levels and clinical outcomes is tenuous, routine measurement is not indicated and should be reserved for special populations, such as patients with renal disease, obese patients, and underweight patients.24

More recently, there has been a major change in the United States Pharmacopeia (USP) monograph of UFH. As a result of the heparin contamination problem encountered from 2007-2009, a new reference standard for heparin and a new test to determine potency were established by the FDA. These changes have resulted in an estimated 10% reduction in anticoagulant activity of UFH, which was validated. Because of this decrease in potency, the intravenous dose of UFH may need to be increased to achieve target aPTT, and more frequent or intensive aPTT monitoring may be required. No dose adjustment is needed for subcutaneous administration of UFH.25

Adverse effects induced by UFH and LMWHs include bleeding, hematoma, early thrombocytopenia, delayed thrombocytopenia with or without white clot syndrome, hyperkalemia, osteoporosis (with prolonged use), and increase in liver enzyme tests (LETs). An increase in LETs may occur in 10% to 30% of patients receiving LMWHs or high molecular weight heparins. However, the increase in LETs seems benign and has not been associated with any cases of hepatic sequelae. Early-onset heparin-induced thrombocytopenia type 1 (HIT-1) manifests with a decrease in platelets of approximately 50,000/mm3. The decrease in platelets is transient and inconsequential. Delayed-onset heparin-induced thrombocytopenia type 2 (HIT-2) is due to the formation of antiplatelet antibodies between days 6 and 12.

If a patient has heparin-dependent antibodies present in plasma from previous heparin exposure, HIT-2 may occur at any time. HIT-2 is dependent on platelet factor-4 binding. These platelet antibodies aggregate and form the basis for the paradoxical heparin-induced white clot syndrome. The white clot syndrome is a medical emergency that may manifest as pulmonary embolism, MI, stroke, renal or hepatic thrombosis, or skin necrosis and gangrene. The diagnosis of HIT-2 is clinical and may be confirmed by several laboratory tests. Clinical diagnosis of HIT-2 includes a significant reduction in the platelet count of greater than 50% or a decrease in the platelet count to less than 100,000/mm3 or both. Ostensibly, the risk of thrombocytopenia is greatest with UFH and lowest with LMWHs; however, LMWHs cannot be administered as an alternative to heparin because of greater than 95% cross-reactivity.

Fondaparinux (Arixtra), a pentasaccharide-selective anti–factor Xa inhibitor agent, does not possess a risk of cross-reactivity and is currently being studied in clinical trials as a treatment modality for HIT-2 and white clot syndrome. Current treatment options for HIT-2 include the direct thrombin inhibitors (DTIs) argatroban, lepirudin (Refludan), and bivalirudin, or selective factor Xa inhibitor fondaparinux.

The antidote for heparin is protamine sulfate. Protamine sulfate is derived from the sperm of mature testes of salmon and related species. Protamine is electropositive and rapidly binds to the electronegative heparin to form salts that have no anticoagulant effect. Protamine also causes a dissociation of heparin–antithrombin III complexes in favor of a heparin-protamine complex. The recommended neutralizing dose of protamine is 1 mg for every 100 U of heparin, up to a total of protamine 50 mg per dose. Protamine should be administered by slow intravenous infusion over at least 1 to 3 minutes to prevent hypotension, bradycardia, or dyspnea. Patients who have previously received protamine-containing insulin, have undergone a vasectomy, or have a known sensitivity to fish or medications derived from fish (calcitonin-salmon, cold water fish oils containing omega-3 fatty acids, and oyster shell–derived calcium supplements) are at an increased risk for experiencing allergic reactions such as anaphylaxis and developing antiprotamine antibodies.24 Excessive protamine may act as an anticoagulant, resulting in bleeding complications; a careful underdosing strategy is suggested. There is no proven method for neutralizing LMWHs. Protamine seems to neutralize approximately 60% of the anti–factor Xa activity of LMWHs. UFH may be the preferred parenteral anticoagulant in patients who are at risk for clinically significant bleeding, such as patients with end-stage renal disease receiving hemodialysis treatments.

Direct thrombin inhibitors

There are four commercially available highly specific DTIs. Desirudin (Iprivasc) is indicated for deep vein thrombosis prophylaxis, bivalirudin (Angiomax) is indicated for unstable angina, and argatroban and lepirudin (Refludan) are indicated for prophylaxis or treatment of thrombosis in patients with HIT-2 and are used for anticoagulation against thromboembolic conditions in patients with or at risk for HIT-2. Argatroban and lepirudin are considered first-line options. DTIs exert their anticoagulant effects by directly inhibiting the effects of thrombin on a sustained fibrin clot. One molecule of a DTI binds to one molecule of thrombin. DTIs are independent of antithrombin III reactions and are not inhibited by platelet factor IV. aPTT is used to monitor the effects of DTIs and is generally maintained at about 1.5 to 2.5 times the upper limit of the control.

The most common adverse effect of DTIs is minor and major hemorrhage. DTIs may cause allergic skin reactions and anaphylactic reactions manifesting with bronchospasm, stridor, and dyspnea. There are no proven antidotes for DTIs. However, there may be a role for recombinant human factor VIIa (rFVIIa; NovoSeven) in DTI bleeding toxicities. rFVIIa is cloned from hamster kidney cells; it is a vitamin K–dependent glycoprotein (molecular mass 50 Da) structurally similar to human plasma–derived factor VIIa. rFVIIa can activate factor IX to IXa and factor X to Xa, converting prothrombin to thrombin and fibrinogen to fibrin, forming a hemostatic plug.

Natural hirudin is produced in trace amounts by the salivary glands of the leech Hirudo medicinalis. Lepirudin is a recombinant hirudin derived from yeast cells. FDA-approved dosing recommendations include an initial bolus dose of 0.4 mg/kg body weight followed by 0.15 mg/kg/hr continuous infusion. Current ACCP guidelines have a slightly different recommendation. The ACCP recommends a starting dose of no more than 0.10 mg/kg/hr and a lower bolus dose of 0.2 mg/kg to be administered only in the setting of life-threatening or limb-threatening thrombosis. These new recommendations were introduced because of the high rates of bleeding associated with the FDA-approved dosing.26

Lepirudin has a half-life of 1.3 hours. aPTT should be obtained about 4 hours after the start of the infusion. Lepirudin is almost exclusively renally eliminated, requiring careful dosing adjustments in mild to moderate renal dysfunction, and it is contraindicated in patients with severe renal impairment. The anticoagulant effects may be enhanced in patients with hepatic disease, and close monitoring in these patients is recommended. The formation of lepirudin–anti-hirudin antibody complexes has been observed in 40% of patients receiving lepirudin and may enhance the effect of lepirudin by delaying its renal elimination. An advantage of lepirudin is its lack of effect on the international normalized ratio (INR) value, facilitating accurate warfarin monitoring and dosing (see the next section for more details on the INR).

Argatroban is a synthetic agent, derived from l-arginine, that reversibly binds to the thrombin active site. It is administered through continuous intravenous infusion. The half-life of argatroban is 30 to 50 minutes. The route of elimination is primarily via the hepatic CYP3A4/5 isoenzyme system, and the potential for drug interactions exists with CYP3A4/5 inhibitors and inducers. There are four argatroban hepatic metabolites; only M1 is active. It is about threefold to fivefold weaker than the parent drug and is present at 0% to 20% relative to the parent. The recommended initial dose of argatroban is 2 μg/kg/min of body weight up to 130 kg. aPTT should be attained 1 to 3 hours after initiation. The dose should be adjusted until steady-state aPTT is 1.5 to 3 times the initial baseline value but does not exceed 100 seconds at a maximum of 10 μg/kg/min. In critically ill patients or patients with hepatic dysfunction, the initial dose of argatroban should be reduced to 0.2 μg/kg/min or 0.5 μg/kg/min. The effects of argatroban are not significantly influenced by renal impairment, and dosage adjustments are unnecessary in this setting.

When used in combination with warfarin, argatroban, especially at doses exceeding 2 μg/kg/min, has the potential to prolong the INR beyond that of warfarin alone. However, argatroban exerts no additional effects on vitamin K–dependent factor Xa activity. Special warfarin dosing considerations are required when concomitantly using argatroban and warfarin.

Warfarin (coumadin)

Warfarin (Coumadin) is an oral anticoagulant indicated for prophylaxis and treatment of venous thrombosis, pulmonary embolism, and thromboembolic complications associated with atrial fibrillation and cardiac valve replacement and as an adjunct in the treatment of coronary occlusion. Warfarin is also used to reduce the risk of death, reinfarction, and thromboembolic events such as stroke or systemic embolization after MI. Warfarin is a racemic mixture; the (S)-isomer has a half-life of 2 days, and the less potent (R)-isomer has a half-life of 1.3 days, and so warfarin is dosed once daily. The full anticoagulant effect of warfarin has a delayed onset of 3 to 5 days, necessitating overlap with a parenteral heparin agent when rapid anticoagulation is preferred.

The initial dose of warfarin should be the expected maintenance dose. Loading doses of warfarin are no longer recommended. A study found that when hospitalized patients were given an average warfarin maintenance dose of 5 mg, the INR was usually greater than or equal to 2 within 4 or 5 days and was associated with less excessive anticoagulation compared with a 10-mg dose.27 Warfarin starting doses of less than 5 mg may be most appropriate in elderly patients. Warfarin interferes with the hepatic synthesis of vitamin K–dependent clotting factors II, VII, IX, and X and endogenous anticoagulant proteins C and S. The time to complete anticoagulation with warfarin is not immediate. Inhibition of coagulation factors begins 12 to 24 hours after administration; however, the complete antithrombotic effects of warfarin may not occur until 5 to 7 days after initiation of therapy.

The INR is the standard for monitoring warfarin therapy. Prothrombin time (PT) as a tool for monitoring warfarin therapy is problematic because thromboplastin reagents vary in their responsiveness to warfarin-induced reduction in clotting factors, a variability that depends on their method of preparation. The INR is a mathematical correction of the results of the one-stage PT that standardizes the reporting of PT determinations worldwide. The INR takes into account the sensitivity of the thromboplastin used in each specific laboratory to determine the PT. The target INR range for warfarin in most clinical scenarios is 2 to 3. The INR should be used exclusively to dose warfarin clinically; however, the PT should be reviewed in conjunction with the INR to aid in detecting laboratory errors in calculation or assay methodology.

Hemorrhage is the most common adverse effect associated with warfarin and ranges from minor to life-threatening major bleeding. Bleeding manifestations may include ecchymoses, petechiae, purpura, melena, hematochezia, hematuria, hemoptysis, hematemesis, epistaxis, or gingival bleeding. Because warfarin inhibits protein C (half-life 8 hours) and protein S (half-life 30 hours), which have shorter half-lives than factors II (half-life 60 hours), IX (half-life 24 hours), and X (half-life 72 hours), there is a risk of a paradoxical hypercoagulability, thrombus formation, and skin necrosis with gangrene. The procoagulant effect of warfarin can be enhanced in patients who have protein C and protein S deficiency. To minimize the immediate procoagulant effect of warfarin, an overlap of 3 to 5 days with a parenteral anticoagulant is warranted. Purple toe syndrome, caused by the release of atheromatous plaque emboli and cholesterol-rich microembolization, occurs approximately 3 to 10 weeks after initiation of coumarin therapy. Purple toe syndrome is reversible and is typically characterized by a purplish or mottled discoloration of the plantar surfaces and sides of the toes that blanches on moderate pressure and fades with elevation of the legs. Oral or parenteral vitamin K1 (phytonadione) may be administered to reverse the anticoagulation effects of warfarin.

Many factors, such as diet, disease states, and drugs, can alter the pharmacologic characteristics and effects of warfarin.28 Patients should be counseled to eat a healthy and consistent diet. An increased intake of vitamin K–containing supplements and foods, such as green leafy vegetables, may result in a reduced anticoagulant response, decreased INR, and subsequently treatment failure such as an embolism. Conversely, abrupt decreases in vitamin K dietary intake may result in an increased anticoagulant response with an increased INR and subsequent risk of hemorrhage. Hepatic disease and, to a lesser extent, renal disease may decrease elimination of warfarin and increase the effects of warfarin. A plethora of drugs can increase or decrease the effects of warfarin. It is prudent to measure the INR frequently when factors that interact with warfarin are added to a patient’s regimen. Table 22-13 lists selected significant warfarin drug interactions.

These agents may increase anticoagulant effect of warfarin by inhibiting hepatic cytochrome P450 isozymes (CYP2C9, CYP3A4, or CYP1A2) involved in its metabolism; risk of bleeding may be increased These agents may increase anticoagulant effect of warfarin by displacement from protein-binding sites (albumin); risk of bleeding may be increased These agents may increase anticoagulant effect of warfarin by inhibiting gastrointestinal vitamin K or by inhibiting platelet aggregation; risk of bleeding may be increased These agents may decrease anticoagulant effect of warfarin by induction of hepatic cytochrome P450 isozymes (CYP2C9, CYP3A4, or CYP1A2) involved in its metabolism; lack of warfarin efficacy and thrombosis may occur These agents may decrease anticoagulant effect of warfarin by various mechanisms; lack of warfarin efficacy and thrombosis may occur

image

NSAIDs, Nonsteroidal antiinflammatory drugs; TMP-SMX, trimethoprim-sulfamethoxazole.

The role of genetic polymorphism in the management of warfarin has been the focus of interest in more recent studies. CYP2C9, which plays an integral role in the metabolism of the S-isomer of warfarin, and VKORC1 (VKOR complex subunit 1), the gene that determines the activity of vitamin K epoxide reductase, both undergo genetic polymorphism leading to variances in warfarin responses between patients. Studies have shown that testing for a patient’s genetic type can lead to decreased major bleeding or thromboembolic events resulting in hospital admission.28 Genetic test kits (e.g., GeneMedRx) are available for purchase but are not covered by insurance plans and can be expensive. Genotyping is still considered a novelty and not routinely used in practice.

Because warfarin is a high-risk medication that can significantly interact with many medications, requires frequent INR monitoring, and is pharmacokinetically challenging to dose, pharmacist-based warfarin clinics with physician supervision and collaboration have become a standard of best practice. More than 1500 such clinics are active in the United States today.

Antiplatelet agents

Aspirin

In platelets, the prostaglandin derivative thromboxane A2 is a major inducer of platelet aggregation and vasoconstriction. Aspirin is hydrolyzed to salicylic acid and inhibits prostaglandin production by acetylating cyclooxygenase, the initial enzyme in the prostaglandin biosynthesis pathway. This inhibition of platelet aggregation lasts for the life of the platelet, which is approximately 7 to 10 days. By inhibiting platelet aggregation, aspirin increases bleeding times. Low doses of aspirin inhibit platelet aggregation, whereas larger doses inhibit cyclooxygenase in arterial walls, which interferes with prostacyclin production. Prostacyclin is a potent vasodilator and inhibitor of platelet aggregation. Lower doses plausibly may be more effective than higher doses in preventing coronary heart disease; however, this has not been proven clinically.

Aspirin has many indications including fever and pain associated with headaches, neuralgias, myalgias, and arthralgias. Antithrombotic indications for aspirin include reducing the risk of thrombosis, such as in the primary and secondary prevention of nonfatal or fatal MI in patients with or without previous MI or unstable angina, and preventing recurrent transient ischemic attacks (TIAs) or stroke. The dose of aspirin for its analgesic, antiinflammatory, and antipyretic effects is considered high dose and may be 325 to 650 mg up to every 4 hours daily as needed. The dose of aspirin for its antithrombotic indications is considered low dose; the range for prevention of MI is 81 to 325 mg daily and for TIA or stroke is 50 to 325 mg daily. Of patients taking aspirin as an antithrombotic, 25% may be genetically prone to aspirin resistance, and higher doses may be necessary to overcome resistance (e.g., 500 mg to 1.5 g daily). Aspirin resistance is best detected via bleeding time tests; however, these tests are not yet validated or standardized, and they are not routinely employed in clinical practice.

Aspirin-induced adverse effects are dose-dependent and include peptic ulcer disease, renal dysfunction, increased blood pressure, tinnitus, pulmonary dysfunction, and bleeding. The risk of clinically significant hemorrhage with aspirin (e.g., gastrointestinal bleeds) is dose-dependent. However, any dose of aspirin carries a risk of major bleeding compared with placebo controls. Patients should be counseled on the signs and symptoms of bleeding, which may include anemia, abnormal bruising, epistaxis, or bleeding of the gums and dizziness and lightheadedness associated with low blood pressure and rapid heart rate. Aspirin, especially at high doses, can induce or exacerbate asthma by inhibiting bronchodilatory prostaglandins (PGE2 and PGI2) and can exacerbate dyspnea in patients with chronic obstructive pulmonary disease. Aspirin is contraindicated in patients who have a history of allergy, especially anaphylaxis to NSAIDs. Patients with rhinorrhea, nasal polyps, and aspirin-induced or NSAID-induced dyspnea are at greatest risk of aspirin-induced or NSAID-induced anaphylaxis. Aspirin should not be administered to children or teenagers with viral infections because of the risk of Reye syndrome.

An important drug-drug interaction between ibuprofen and aspirin has been identified. Ibuprofen interferes with aspirin access to the platelet serine-binding site and inhibits the pharmacologic effect of aspirin. This drug interaction occurs during single ingestion when ibuprofen is administered before aspirin or with long-term use of ibuprofen and aspirin regardless of whether ibuprofen is administered before or after aspirin. Diclofenac (Voltaren) and celecoxib (Celebrex) do not seem to interact with aspirin; other NSAIDs have not been studied. The combination of aspirin and NSAIDs may lead to a high risk of life-threatening gastropathy, especially in elderly patients or patients using concomitant antithrombotic agents. Aspirin and NSAIDs inhibit gastrointestinal vasodilatory prostaglandins (PGE2 and PGI2), increasing the accumulation of aggressive factors (acid) and decreasing the supply of defensive factors (sodium bicarbonate). Patients should be immediately placed on gastropathy prophylaxis with proton pump inhibitors (omeprazole, pantoprazole, esomeprazole, or lansoprazole) or misoprostol (Cytotec).

Dipyridamole

Dipyridamole is a vasodilator and platelet adhesion inhibitor. It has been postulated that patients with prosthetic heart valves have abnormally shortened platelet survival time. Dipyridamole lengthens the abnormally shortened platelet survival time in a dose-dependent manner. The primary effect of dipyridamole is to inhibit cGMP-specific phosphodiesterase, increasing cGMP levels and augmenting the effects of nitric oxide. Dipyridamole weakly inhibits red blood cell, endothelial cell, and platelet uptake of the platelet activity inhibitor adenosine and inhibits the formation of thromboxane A2; this effect occurs in a dose-dependent manner (0.5 to 1.9 μg/mL). This uptake inhibition results in dipyridamole inhibiting platelet function by inhibiting cyclic adenosine 3′,5′-monophosphate (cAMP)–specific phosphodiesterase, which leads to increased cellular concentrations of cAMP within platelets, preventing platelet aggregation by stimuli such as collagen and ADP. Dipyridamole does not alter PT levels but can increase the platelet bleeding time.

Dipyridamole (Persantine) is indicated only as an adjunct to warfarin in the prevention of postoperative thromboembolic complications of cardiac valve replacement. Intravenous dipyridamole, occasionally used for cardiac exercise stress testing, may decrease blood pressure and increase heart rate and cardiac output; this effect is generally not seen with the oral dosage form. Dipyridamole is eliminated via hepatic conjugation and glucuronidation; it does not undergo hepatic cytochrome P450 elimination. Dipyridamole has a weak metabolite and undergoes negligible renal elimination. The half-life of dipyridamole is 13 hours. Adverse reactions are transient and include headache, dizziness, hypotension, and abdominal distress. Rarely, dipyridamole has aggravated angina symptoms; the intravenous form has precipitated acute myocardial ischemia. Dipyridamole can potentiate the effects of intravenous adenosine, causing fatal asystole or sustained ventricular tachycardia; a decreased dose of adenosine should be used when treating paroxysmal supraventricular tachycardias.

Aggrenox is a combination gelatin capsule containing 200 mg of extended-release dipyridamole with 25 mg of aspirin and is indicated to reduce the risk of stroke for patients who have had TIAs or complete ischemic strokes. Steady-state dipyridamole peak and trough plasma levels are 2 μg/mL and 0.5 μg/mL, allowing for dipyridamole to achieve its effects on cAMP and cGMP throughout the dosing interval; this is not likely to occur with prompt-release dipyridamole. Dipyridamole requires an acidic environment for gut absorption; Aggrenox contains tartaric acid, allowing for maximal bioavailability in patients who have gut hypochlorhydria or achlorhydria (e.g., elderly patients).

The second European Stroke Prevention Study (ESPS-2) showed that dipyridamole modified-release formulation, 200 mg given twice daily, is effective in the secondary prevention of stroke and TIA compared with placebo and that coadministration with aspirin, 25 mg twice daily, provides additional benefit.28 ESPS-2 showed that the relative risk reduction for stroke with aspirin administration compared with placebo was 18.1% (P = .013); for modified-release dipyridamole, the relative risk reduction was 16.3% (P = .039); and with the combination, it was 37% (P < .001). Aggrenox must not be substituted by prompt-release dipyridamole and aspirin. The recommended dosage of Aggrenox is one capsule twice daily, swallowed whole. Because of increased risk of headache, it may be advisable to start with one capsule daily at bedtime up to 1 week until tolerated before increasing the dose to twice daily.

Clopidogrel (plavix)

Clopidogrel (Plavix) is a prodrug thienopyridine derivative platelet aggregation inhibitor that interferes with platelet membrane function by inhibiting ADP-induced platelet-fibrinogen binding and subsequent platelet-platelet interactions. Indications for clopidogrel include the reduction of atherosclerotic events in patients with a history of MI, stroke, or peripheral arteriolar disease and acute coronary syndrome (ACS) regardless of whether a patient is managed medically, by percutaneous coronary intervention (PCI), or by coronary artery bypass grafting. Clopidogrel is slightly more effective than aspirin in reducing the combined risk of ischemic stroke, MI, or vascular death in patients with atherosclerotic vascular disease.29 For patients with ACS, clopidogrel plus aspirin was found to be superior to aspirin alone in reducing composite endpoints of MI, stroke, and cardiovascular death. Clopidogrel has not shown superiority to aspirin for stroke prophylaxis except in patients with peripheral vascular disease.

Clopidogrel is extensively metabolized by the liver; its metabolites are eliminated equally via the kidneys and the feces. The half-life of clopidogrel metabolites is 8 hours; steady state is reached in 3 to 7 days. The onset of action of clopidogrel can be seen in 2 hours. The average platelet inhibition seen with clopidogrel is between 40% and 60%. Platelet aggregation and bleeding time return to normal within 5 days after clopidogrel discontinuation. The dose of clopidogrel in ACS is a 300-mg loading dose followed by 75 mg once daily (plus aspirin). A higher loading dose of 600 mg is recommended for patients undergoing PCI. For the prevention of cardiovascular events such as ACS, stroke, and peripheral arterial disease, the dose is 75 mg of clopidogrel once daily.

Because clopidogrel is a prodrug, it must undergo a two-step hepatic conversion to be activated. Only about 15% of the drug is converted to its active form, mainly through CYP2C19 and CYP3A4. Several trials have evaluated the possible interactions between clopidogrel and other drugs that can inhibit or competitively bind to these same isoenzymes, potentially inhibiting the conversion of clopidogrel to its active form and rendering the drug ineffective. Proton pump inhibitors (PPIs), which are commonly prescribed for prophylaxis of stress ulcer, gastrointestinal reflux disease, and prophylaxis of gastrointestinal bleeding, are metabolized by CYP2C19 at varying degrees. Studies have reported that there is a significant increase in clinical event rates (e.g., MI, death) or greater platelet reactivity with concurrent use of clopidogrel and a PPI. Pantoprazole (Protonix) is the only PPI that does not undergo CYP2C19 metabolism and may potentially be devoid of this interaction, but more studies need to be conducted to confirm this. Other medications such as cimetidine, etravirine, felbamate, fluconazole, fluvoxamine, fluoxetine, ketoconazole, voriconazole, and ticlopidine should also be avoided because they can reduce antiplatelet activity of clopidogrel.

Statins or 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase inhibitors have also been an area of interest with regard to clopidogrel drug interactions. Similar to clopidogrel, statins are CYP3A4 substrates. In vivo studies that examined the degree of platelet inhibition by clopidogrel in patients taking a statin have shown that there is a significant decline in platelet inhibition with the use of statins. However, no large clinical trials have shown that this interaction can lead to negative clinical outcomes. Despite the lack of clinical trials evaluating the interaction between statins and clopidogrel, caution is advised when combining these two medications. If statin therapy is required, pravastatin (Pravachol) or rosuvastatin (Crestor) should be considered because these drugs do not undergo CYP3A4 metabolism and potentially are devoid of any significant drug interactions.30

Testing for genetic polymorphism has also received considerable emphasis in the FDA boxed warning for clopidogrel. Although genetic polymorphism for CYP2C19 has been shown in several studies to reduce antiplatelet activity and increase major adverse cardiac events (MACEs), prospective studies show clinical efficacy of personalizing antiplatelet therapy based on genotype analysis. The recommendation issued by the FDA to clinicians is to consider alternative treatment strategies, such as combining clopidogrel with cilostazol or using high-dose clopidogrel (600 mg loading dose followed by 150 mg daily for ACS) in patients identified as poor CYP2C19 metabolizers. The efficacy and safety of these approaches remain uncertain.31 Alternatively, one may consider switching to another antiplatelet agent, such as prasugrel, which does not undergo CYP2C19 metabolism.

The most common adverse effect of clopidogrel is hemorrhage, manifesting with purpura and epistaxis; other adverse effects include headaches and dizziness, abdominal pain and diarrhea, and rash and pruritus. In contrast to ticlopidine, clopidogrel has a lower incidence of rash, gastrointestinal disturbances, neutropenia, and thrombotic thrombocytopenic purpura (TTP), and cholestatic jaundice has not been reported with clopidogrel.

Ticlopidine (ticlid)

Ticlopidine (Ticlid), a thienopyridine, is a platelet aggregation inhibitor that interferes with platelet membrane function by inhibiting ADP-induced platelet-fibrinogen binding and subsequent platelet-platelet interactions. The effect of ticlopidine on platelet function is irreversible and lasts for the life of the platelet. Ticlopidine is indicated for stroke. In the Ticlopidine Aspirin Stroke Study (TASS), the ticlopidine group had a 21% greater relative risk reduction for stroke compared with the aspirin group and a 9% greater reduction in stroke, MI, or vascular death at 3 years.31 The Canadian-American Ticlopidine Study (CATS) showed that ticlopidine reduced the relative risk of stroke, MI, or vascular death by 30% compared with placebo (10.8%; P = .006).32 The half-life of ticlopidine is 14 hours; however, with repeat dosing it approaches 4 to 5 days. Steady state is achieved within 14 to 21 days. Platelet aggregation is inhibited 50% within 4 days and 60% to 70% within 10 days. Platelet aggregation and bleeding time return to normal within 14 days after ticlopidine discontinuation. Ticlopidine is extensively metabolized by the liver; active metabolites have not been elucidated.

Because of the risk of life-threatening blood dyscrasias such as TTP and neutropenia and agranulocytosis, ticlopidine is a refractory agent reserved for patients who are intolerant or allergic to aspirin or clopidogrel or failed therapy with these agents. The onset of TTP occurs after 3 to 4 weeks of therapy, and the onset of neutropenia occurs after 4 to 6 weeks of therapy. TTP rarely occurs after 3 months of therapy. The incidence of TTP may be 1 case in every 1600 to 4000 patients. Signs and symptoms of TTP include microangiopathic hemolytic anemia (schistocytes on peripheral smear), purpura, petechiae, pallor, renal dysfunction, fever, weakness, difficulty speaking, seizures, jaundice, and dark or bloody urine. With proper detection, ticlopidine discontinuation, and management by plasmapheresis, 80% of patients survive. Because of the risk of ticlopidine-induced TTP and neutropenia, patients should have a complete blood count monitoring neutrophils, platelets, and hemoglobin and hematocrit biweekly for the first 3 months.

Rarely, ticlopidine can cause thrombocytopenia not induced by TTP. Ticlopidine may cause gastrointestinal disturbances such as diarrhea, nausea, and vomiting in one-third of patients. Similar to clopidogrel, it does not directly cause peptic ulcer disease. Rare cases of rash that may progress to Stevens-Johnson syndrome and cholestatic jaundice have occurred with ticlopidine use. Ticlopidine may persistently increase total cholesterol levels by 10%.

Prasugrel (effient)

Prasugrel (Effient) is a prodrug thienopyridine derivative platelet aggregation inhibitor that interferes with platelet membrane function by inhibiting ADP-induced platelet-fibrinogen binding and subsequent platelet-platelet interactions. When prasugrel is compared with other agents in the same class, it has a very limited scope; it is indicated only for the prevention of thrombosis in patients with ACS undergoing PCI. Prasugrel in combination with aspirin decreases nonfatal MI slightly more than clopidogrel in combination with aspirin but with an increased risk of bleeding. Prasugrel is contraindicated in patients who have had history of a TIA or stroke. In patients older than 75 years or in patients who weigh less than 60 kg, prasugrel was shown to have a greater risk of bleeding, which outweighs its benefit.33 Prasugrel is extensively metabolized by hydrolysis in the liver followed by CYP3A4 and CYP2D6; its metabolites are eliminated via the kidneys and feces. The half-life of the active metabolite of prasugrel is 7 to 8 hours. The onset of action of prasugrel can be seen in 30 minutes. The average platelet inhibition observed with prasugrel is 50% to 80%. Platelet aggregation and bleeding time return to normal approximately 5 to 9 days after prasugrel discontinuation. The dose of prasugrel in ACS is a 60-mg loading dose followed by 10 mg once daily (plus aspirin). In patients who weigh less than 60 kg, a decreased dose of 5 mg is recommended, although there have not been any clinical trials to support this practice.

The most common adverse effect of prasugrel is hemorrhage, which is greater than that observed with clopidogrel. Other adverse effects are similar to clopidogrel. A higher incidence of colonic neoplasm was seen during the trial for its approval.34,35

Cilostazol (pletal) and pentoxifylline (trental)

Cilostazol (Pletal) is a quinolinone derivative that selectively and reversibly inhibits cellular phosphodiesterase III by increasing the levels of cAMP, resulting in vasodilation and inhibition of platelet aggregation. Cilostazol is indicated for intermittent claudication in patients with peripheral arterial disease. Cilostazol allows for increased walking distances and improves symptoms and quality of life in patients with intermittent claudication. The clinical benefits may not be noticed for at least 2 to 4 weeks, and may take 12 weeks. The only alternative to cilostazol for intermittent claudication is pentoxifylline (Trental); however, it has been proven inefficacious. Pentoxifylline is a xanthine agent with rheologic properties that decrease blood viscosity and improve erythrocyte flexibility.

Cilostazol is associated with a high incidence of transient adverse effects, such as headache, diarrhea, dizziness, and palpitations. In patients with heart failure, oral phosphodiesterase inhibitors such as milrinone have been associated with increased mortality resulting from arrhythmias; cilostazol and several of its metabolites are contraindicated in patients with heart failure and should be used prudently in patients with CAD. Cilostazol has been associated with increases in heart rate and reductions in P–R, QRS, and Q–T intervals on ECG. In the dog model, cilostazol has been associated with cardiac lesions and endocardial hemorrhage, similar to toxicities noted with milrinone; the risk of developing cardiac lesions with long-term cilostazol use is unknown. Cilostazol is a CYP3A4 and CYP2C19 substrate and has been associated with significantly elevated levels when combined with the CYP3A4 inhibitors ketoconazole, diltiazem, and erythromycin. Smokers exhibited 20% lower levels of cilostazol. Cilostazol is dosed at 100 mg twice daily; the dose should be reduced in the presence of CYP3A4 inhibitors to 50 mg twice daily. Food increases the bioavailability of cilostazol by 90%; cilostazol should be administered on an empty stomach to circumvent this interaction. Grapefruit juice inhibits gut CYP3A4 and may increase plasma cilostazol levels and should be avoided during cilostazol use.

Glycoprotein iib/iiia inhibitors

GP IIb/IIIa inhibitors are indicated for the treatment of patients with ACS—unstable angina or non–ST segment elevation acute MI—and patients who are medically managed and patients undergoing PCI. Abciximab (ReoPro) is the GP IIb/IIIa inhibitor of choice for PCI. The management of unstable angina or non–ST segment elevation acute MI includes the use of aspirin, heparin, and a GP IIb/IIIa inhibitor. This combination has led to a decrease in the composite endpoints of new MI or death.36 In patients who are being managed medically for a non–ST segment elevation ACS, the use of GP IIb/IIIa inhibitors has been marginalized to patients with moderate to high risk based on a risk assessment score and continued ischemia or patients with diabetes. GP IIb/IIIa inhibitors are administered via continuous intravenous infusions. Oral formulations of GP IIb/IIIa inhibitors failed to display efficacy in clinical trials and are unavailable.

The most common adverse effect reported during therapy was bleeding. The incidence of major bleeding manifesting as gastrointestinal, genitourinary, or intracranial hemorrhage with the three-drug combination was only slightly greater than with aspirin and heparin alone, illustrating the safety of the GP IIb/IIIa inhibitors. Although minor bleeding with GP IIb/IIIa inhibitors is common (10%), it is generally inconsequential. Because these agents may cause thrombocytopenia, daily platelet, hemoglobin, and hematocrit monitoring is required. Abciximab has been implicated as a cause of immune-mediated thrombocytopenia in 5% of patients. Table 22-14 presents the pharmacologic characteristics of GP IIb/IIIa inhibitors.

TABLE 22-14

Characteristics of Glycoprotein (GP) IIb/IIIa Inhibitors

  ABCIXIMAB (REOPRO) TIROFIBAN (AGGRASTAT) EPTIFIBATIDE (INTEGRILIN)
Common uses Adjunct to PCI Management of ACS, medically or with PCI Management of ACS, medically or with PCI; adjunct to PCI
Pharmacology Chimeric human-murine monoclonal antibody Fab fragment GP IIb/IIIa inhibitor Nonpeptide GP IIb/IIIa inhibitor Cyclic heptapeptide GP IIb/IIIa inhibitor
Origin Antibodies from immunized mice Chemically derived Active component of snake venom peptides
Binding to platelets Irreversible Reversible Reversible
Elimination half-life 30 minutes 2 hours 2.5 hours
Platelet function recovery Approximately 48 hours Approximately 4 hours Approximately 4 hours
Elimination Renal, lymphatic system 65% renal, 25% biliary 50% renal, 30% metabolized in plasma into amino acids

image

ACS, Acute coronary syndrome; PCI, percutaneous coronary intervention.

Thrombolytic agents

Thrombolytics are indicated for the management of pulmonary embolism, ischemic stroke, and acute ST segment elevation MI—the most extensive and life-threatening type of heart attack. Thrombolytics reduce the incidence of heart failure and death associated with acute MI and restore coronary blood flow by dissolving the thrombus, limiting the extent of ischemia and necrosis. Thrombolytics convert plasminogen to plasmin. Subsequently, the proteolytic enzyme plasmin initiates clot lysis and produces FDPs.

For the treatment of ST segment elevation ACS manifesting with at least 1 mm of ST segment elevation in two or more contiguous ECG leads, all available thrombolytics (streptokinase, alteplase, reteplase, and tenecteplase) are indicated; however, streptokinase is considered a second-line agent because of its lack of fibrin specificity. Eligible patients should receive thrombolytic therapy within 12 hours of symptom onset; however, benefit can be realized for 24 hours. Thrombolytics are preferred to primary PCI when patients present within 3 hours of symptom onset and the door to primary PCI time would be greater than 90 minutes. For acute massive pulmonary embolism, alteplase is the only thrombolytic indicated. Alteplase is reserved for patients with acute massive pulmonary embolism who present with symptoms within 2 weeks but optimally within 5 days. Alteplase is the only thrombolytic indicated for the management of acute ischemic stroke for patients who present within 3 hours of symptom onset and no later than 6 hours after onset.37 The use of thrombolytics is often precluded because of their extensive list of contraindications. The absolute and relative contraindications for the use of thrombolytics are listed in Box 22-2.

The most common adverse effect associated with these agents is major and minor bleeding. Major bleeding includes gastrointestinal, genitourinary, respiratory tract, retroperitoneal, and intracranial hemorrhage. Minor bleeding often manifests as superficial or surface bleeding as a result of arterial punctures and surgical intervention. Thrombolytic-induced hemorrhagic stroke in patients older than 75 years occurs more often with alteplase than with streptokinase. Patients older than 75 years should receive streptokinase rather than alteplase. Alteplase and tenecteplase are known to be fibrin specific because they promote the conversion of plasminogen into plasmin in the presence of clot-bound fibrin only, with limited systemic proteolysis. The increased fibrin specificity is believed to induce less extensive systemic depletion of clotting factors such as fibrinogen and plasminogen. The clinical relevance of thrombolytic fibrin specificity has not been elucidated. Thrombolytics have rarely been associated with cholesterol embolization manifesting as purple toe syndrome, livedo reticularis, acute renal failure, gangrene, MI, bowel infarction, stroke, and rhabdomyolysis. When thrombolytics are used for ACS, they can cause reperfusion arrhythmias manifesting as bradycardia or ventricular tachyarrhythmias. Table 22-15 presents the pharmacologic properties of thrombolytic agents.38

TABLE 22-15

Pharmacologic Properties of Thrombolytic Agents

  STREPTOKINASE ALTEPLASE (rtPA) RETEPLASE (rPA) TENECTEPLASE (TNK-tPA)
Brand name Streptase Activase Retavase TNKase
Source Streptococcal culture Recombinant DNA technology using heterologous mammalian tissue culture Recombinant DNA technology using Escherichia coli Recombinant DNA technology using Chinese hamster ovary cells
Common uses Pulmonary embolism, deep vein thrombosis, peripheral arterial occlusion, clearance of occluded central venous access devices, ST segment elevation Pulmonary embolism, stroke, clearance of occluded central venous access device, ST segment elevation Myocardial infarction, ST segment elevation Myocardial infarction, ST segment elevation
Type of agent Bacterial proactivator Tissue plasminogen activator Tissue plasminogen activator Tissue plasminogen activator
Plasma half-life (min) 12-18 2-6 13-16 90-130
Fibrinolytic activation Systemic Systemic Systemic Systemic
Antigenic Yes No No No
Fibrin specific + +++ ++ ++++
Systemic bleeding risk +++ ++ ++ +
ICH risk + ++ ++ ++

image

ICH, Intracranial hemorrhage; rPA, recombinant plasminogen activator; rtPA, recombinant tissue-type plasminogen activator; TNK, tenecteplase; tPA, tissue-type plasminogen activator.