Pharmacologic Options for Treatment of Ischemic Disease




Organic Nitrates


Overview


Nitroglycerin (glyceryl trinitrate [GTN]) was first synthesized in 1847 by Ascanio Sobrero, who described a “violent headache” upon self-administration of a “minute quantity” of the drug. A number of reports of the therapeutic effects of GTN in the latter half of the nineteenth century included those of Field, Brunton, and Murrell. Although sublingual GTN has been commonly used for more than a century to treat acute attacks of angina, the development of organic nitrates with sustained activity was limited by their poor oral bioavailability. Eventually, this difficulty was overcome with transdermal formulations of GTN and the development of long-acting oral nitrate preparations, including isosorbide dinitrate, isosorbide-5-mononitrate, erythrityl tetranitrate, and pentaerythritol tetranitrate. Today, the organic nitrates continue to play an important role in the management of both angina and congestive heart failure (CHF).


Mechanisms of Action


Organic nitrates are prodrugs that must undergo enzymatic denitrification to mediate their pharmacodynamic effects ( Figures 7-1 and 7-2 ). In 1977, Murad first suggested that nitric oxide (NO) mediated the effects of GTN. Since that time, it has generally been accepted that all the organic nitrates exert their effects via release of NO or some NO-containing moiety. As the understanding of NO biology grew and the importance of decreased NO bioavailability in cardiovascular (CV) disease was recognized, it was postulated that the organic nitrates could supplement endogenous NO with favorable biologic effects. Despite its appeal, this hypothesis has never been formerly tested, and no available evidence suggests that exogenous NO donors favorably modify the natural history of CV disease.




FIGURE 7-1


Pathways of organic nitrate bioactivation in vascular cells. ALDH2, aldehyde dehydrogenase 2; cGMP, cyclic guanosine monophosphate; cGK-I, cGMP dependent protein kinase-I; Cyt Ox, cytochrome oxidase; ER, endoplasmic reticulum; GDN, glycerol dinitrate; GMN, glycerol mononitrate; GTN, glyceril trinitrate (nitroglycerin); ISDN, isosorbide dinitrate; ISMN, isosorbide mononitrate; NO, nitric oxide; PEDN, pentaerythrityl dinitrate; PEMN, pentaerythrityl mononitrate; PETN, pentaerythrityl tetranitrate; PETriN, pentaerythrityl trinitate; sGC, soluble guanylate cyclase.



FIGURE 7-2


Molecular mechanisms of nitrate tolerance. ALDH2, aldehyde dehydrogenase; AT-II, angiotensin II; BH2, dihydrobiopterin; BH4, tetrahydrobiopterin; cGMP, cyclic guanosine monophosphate; ET-1, endothelin-1; GMP, guanosine monophosphate; GTN, glyceril trinitrate (nitroglycerin); GTP-CH, guanosine triphosphate cyclohydrolase; NOS, nitric oxide synthetase; PDE, phosphodiesterase; PGI, prostacyclin; PKC, protein kinase C; RAAS, renin-angiotensin-aldosterone system; NADPH-Ox, nicotinamide adenine dinucleotide phosphate oxidase; sGC, soluble guanylate cyclase.


The exact mechanism of nitrate biotransformation (denitrification) was the subject of debate for several decades. Multiple enzymatic candidates were proposed, including cytochrome P450 (CYP), endothelial NO synthase, and glutathione transferase. Interest in defining the denitrification pathway was intense because it was believed that the development of abnormalities in this process might explain the loss of nitrate effects during sustained therapy, a phenomenon termed nitrate tolerance. More recent studies have proposed a role for mitochondrial aldehyde dehydrogenase type 2 (mALDH-2) in the biotransformation of GTN. The role of this enzyme in the activation of GTN supported the dependence of GTN-induced cyclic guanosine monophosphate (cGMP) production, based on the presence of this enzyme and the fact that specific antagonists of mALDH-2 inhibit the vasoactive actions of GTN. The relevance of this biotransformation pathway in humans is supported by observations of reduced hemodynamic responses to GTN in Asian subjects, who are genetically deficient in the enzyme aldehyde dehydrogenase. Importantly, the function of this biotransformation pathway is consistent with a number of observations concerning nitrate tolerance (see the discussion below). Although the discovery of the mALDH-2 biotransformation pathway provided important new insights concerning the actions of GTN, many unanswered questions remain. Most importantly, the denitrification of other organic nitrates, including isosorbide dinitrate and isosorbide-5-mononitrate, does not depend on mALDH-2. These observations emphasize that our knowledge of these processes remains incomplete.


Following organic nitrate bioactivation, and despite uncertainties concerning the exact nature of the NO moiety derived, there is activation of soluble guanylate cyclase and increased cGMP synthesis. The subsequent increase in the bioavailability of cGMP triggers a molecular cascade that mediates vasorelaxation through multiple pathways, which lead to a reduction in intracellular Ca 2+ concentrations, including activation of protein kinases involved in the regulation of intracellular Ca 2+ levels, such as the sarcoplasmic Ca 2+ -ATPase. NO donors, as with endogenous NO, appear to have multiple biologic effects, including thiol modification, regulation of mitochondrial respiration, modulation of K + channel activity, and protein nitration, although less evidence is available concerning the therapeutic relevance of these effects. As discussed below, these diverse biochemical responses to the organic nitrates appears to be responsible for their recently described nonhemodynamic effects.


Pharmacokinetics


Organic nitrates are available in a variety of formulations with differing routes of administration. GTN undergoes hepatic and intravascular metabolism with a half-life of approximately 1 to 4 minutes with biologically active dinitrate metabolites that have a half-life of approximately 40 minutes. GTN is very effective when given by the sublingual and transdermal routes. Transdermal administration of GTN is the only method of administration that provides a clinically effective long-acting form of GTN. When given orally, first-pass metabolism of GTN is extensive. Oral GTN is available for the therapy of angina, but no evidence of clinical efficacy exists.


Although it provides hemodynamic and antianginal effects, isosorbide dinitrate is rapidly metabolized, with a plasma half-life of approximately 40 minutes. Its major metabolites, isosorbide-2-mononitrate and isosorbide-5-mononitrate, are both biologically active, with half-lives of approximately 2 and 4 hours, respectively. Isosorbide-5-mononitrate does not undergo first-pass hepatic metabolism and is completely bioavailable. Both of these nitrates are available in sustained-release preparations; the sustained, phasic-release form of isosorbide-5-monitrate is the most popular, with once-daily dosing and favorable pharmacokinetics that avoid tolerance. Although not prescribed in North America, both erythrityl tetranitrate and pentaerythritol tetranitrate are used in certain parts of the world for the treatment of angina.


Pharmacodynamic Effects


The organic nitrates are potent vasodilators, whose vascular effects vary widely in different distributions of the vasculature ( Figure 7-3 ). They have potent effects in the venous capacitance bed and reduce ventricular volume and preload. They also dilate conduit arteries, and at the doses used clinically, they have no effect on peripheral vascular resistance. The nitrates dilate epicardial coronary arteries but have little or no effect on the coronary resistance vessels. In patients with coronary artery disease (CAD), nitrates can dilate coronary stenoses and collateral vessels, which can improve and redistribute coronary blood flow. Because they do not reduce coronary vascular resistance, nitrates avoid the risk myocardial ischemia because of coronary steal, which can occur with arteriolar dilators, such as dipyridamole and short-acting dihydropyridines (DHPs), in which coronary blood flow is diverted away from areas of ongoing ischemia. Therefore, the nitrates possess a unique combination of vascular effects that can favorably affect the mismatch between myocardial oxygen supply and demand in patients with CAD.




FIGURE 7-3


Antianginal effects of acutely administered glyceryl trinitrate ( GTN ; nitroglycerin). LVEDP, left ventricular end-diastolic pressure.


Side Effects of Organic Nitrates


Headaches are common during nitrate therapy and are generally most pronounced early after initiation of therapy ( Table 7-1 ). In some patients, the headache diminishes over a few days, but not uncommonly, the nitrate must be discontinued. Hypotension can occur with all nitrates but is more common when nitrates with a rapid onset of action are used, such as sublingual GTN or short-acting isosorbide dinitrates. Many patients experience dizziness, presyncope, or even syncope on initial exposure to sublingual GTN or initial doses of isosorbide dinitrate. Symptomatic hypotension is less common after transdermal GTN administration. In general, patients taking their first dose of nitrates should sit or lie down during administration if necessary. In the case of isosorbide dinitrate, the dose should be uptitrated over several days, starting with the 10-mg dose. Reduction in dose or a change of agent should be considered when such symptomatic hypotension occurs. Other side effects are uncommon. With transdermal GTN, some patients develop marked erythema and some local edema at the site of the application of the transdermal preparation. This likely represents a marked response to local hyperemia, but in some patients, it may represent a local allergic reaction to the preparation itself. In some this reaction is troublesome enough that the transdermal preparation must be discontinued. Although rare, methemoglobinemia has been reported after high-dose intravenous GTN therapy.



TABLE 7-1

Nitrate Preparations, Routes of Administration, and Dosing Strategies































































DRUG ROUTE DOSE RANGE FREQUENCY
Treatment of Anginal Attacks
Nitroglycerin tablets
Nitroglycerin spray
Nitroglycerin buccal tablets
Sublingual
Sublingual
Buccal
0.3-0.6 mg
0.4-0.8 mg
1-3 mg
1-3 times
1-3 times
Once
Prevention of Anginal Attacks
Nitroglycerin tablets
Nitroglycerin spray
Sublingual
Sublingual
0.3-0.6 mg
0.4-0.8 mg
2-5 minutes before activity
Nitroglycerin buccal tablets Buccal 1-3 mg 2-5 minutes before activity or three times daily; tablet is removed overnight
Nitroglycerin SR Oral 2.6-10.4 mg Two to three times daily
Nitroglycerin patch Transdermal 0.2-0.8 mg/h Once daily, 12-h patch-free interval
Isosorbide dinitrate SF Sublingual 2.5-10 mg 5-10 min before activity
Isosorbide dinitrate SF Oral 10-45 mg 3 times daily, 14-h tablet-free interval
Isosorbide dinitrate SR Oral 20-80 mg Once or twice daily
Isosorbide-5-mononitrate SF Oral 10-20 mg Twice daily, 7 h between doses
Isosorbide-5-mononitrate SR Oral 30-240 mg Once daily

SF, standard formulation; SR, sustained-release formulation that provides therapeutic plasma concentrations for 12 hours.


Clinical Efficacy of Organic Nitrates


Sublingual Nitrates


Sublingual GTN represents a classic therapy for the treatment of acute attacks of angina ( Table 7-2 ; also see Figure 7-3 ). Whether given as a tablet or spray, it has a rapid onset of action that offers prompt symptomatic relief. Historically, sublingual GTN was often prescribed as a prophylactic therapy, taken by the patient before activity that would generally lead to anginal symptoms. Given in this manner, it significantly increases exercise capacity, a finding that has now been confirmed in clinical trials. In select patients, this can be a very effective way to improve symptoms and quality of life, when other approaches to the prevention of angina are not effective. Sublingual isosorbide dinitrate is also available. Although not commonly used, it can both treat and prevent angina in select patients.



TABLE 7-2

Side Effects of Organic Nitrates
























DRUG ROUTE SIDE EFFECTS COMMENTS



  • Nitroglycerin



  • Isosorbide dinitrate

Sublingual


  • Headache



  • Postural hypotension



  • Syncope

Dose reduction may be required



  • Nitroglycerin



  • Isosorbide dinitrate



  • Isosorbide-5-mononitrate



  • Erythrityl tetranitrate



  • Pentaerythritol tetranitrate

Oral


  • Headache



  • Postural hypotension



  • Syncope



  • Nausea




  • Headache and postural lightheadedness often resolve after several days of therapy



  • Resolution of headache does not necessarily mean loss of efficacy

Nitroglycerin Transdermal


  • Same as for oral nitrates



  • Skin erythema and inflammation at site of patch application




  • Initiate treatment with small doses and increase as necessary



  • Vary application site



Long-Acting Nitrates


A classic pharmacodynamic characteristic of the organic nitrates is the phenomenon of tolerance. It has been repeatedly demonstrated that long-acting nitrates are effective in angina, improving exercise duration and reducing the frequency of anginal attacks if given using dosing intervals or formulations that allow for a low or nitrate-free period during the day. Isosorbide-5-mononitrate in a phasic-release formulation that provides effective plasma concentrations during the day but low concentrations during the night is effective in the therapy of exertional angina. In some countries, the organic nitrate pentaerythritol tetranitrate is also prescribed for the therapy of angina. This nitrate appears to have some unique biochemical properties that make is less susceptible to tolerance. Unfortunately, few data are available to document its antianginal effects.


Congestive Heart Failure


The administration of nitrates in patients with CHF has potent and favorable hemodynamic effects. When given acutely, nitrates can dramatically lower filling pressure without adverse effects on systemic blood pressure. In acutely ill patients with markedly elevated filling pressures, sublingual or intravenous GTN can be particularly useful. Although they have not been clearly demonstrated to improve clinical outcome, organic nitrates are generally believed to be safe and effective in the relief of symptoms in patients with acute decompensated heart failure. In patients with acute heart failure and active ischemia, organic nitrates can be the therapy of choice. They are also effective in the therapy of chronic heart failure.


Approximately 25 years ago, the combination of isosorbide dinitrate and hydralazine was the first drug regimen shown to reduce mortality rate in chronic CHF. In 2004, the African-American Heart Failure trial (A-HeFT) documented a favorable effect of this drug combination in African Americans using a twice-daily dosing formulation that combined isosorbide dinitrate and hydralazine. This combination is indicated therapy in African Americans with chronic heart failure as a result of systolic dysfunction, and it is useful as an adjunct therapy in other populations with chronic CHF.


Other Nitrate Indications


Nitrates are also useful in the management of unstable angina and acute myocardial infarction (MI). In patients with acute symptomatic ischemia, nitrates can be extremely effective. Sublingual GTN is often used, but intravenous (IV) and transdermal formulations also have a role. In this setting, beyond their effects on loading conditions, their mechanism of action likely includes their ability to dilate and prevent constriction of epicardial coronary arteries, thus improving coronary blood flow. Furthermore, the potential antiplatelet effects of GTN may play a role in this situation. The question of tolerance in this setting is controversial, although recent evidence suggests that tolerance may not develop to conduit artery dilation and may not develop as rapidly in this situation compared with other clinical settings.


Nitrate Tolerance


When given acutely, nitrates have potent hemodynamic and therapeutic effects. As long-acting nitrates were developed for clinical use, questions arose concerning their potency. Early investigations questioned the efficacy of the oral isosorbide dinitrate, as first-pass metabolism was felt to be complete, because portal vein administration in animals had no hemodynamic effects. Subsequent studies in patients with CAD refuted these findings because oral isosorbide dinitrate clearly produced significant hemodynamic and antianginal effects. However, later investigations confirmed that the clinical effects of nitrates were lost rapidly during sustained therapy, documenting that the phenomenon of nitrate tolerance is a significant clinical problem. Tolerance develops in response to all nitrates, although evidence suggests that it is not as prominent with pentaerythritol tetranitrate. When nitrates are administered using dosing regimens that lead to significant plasma concentrations throughout a 24-hour period, their hemodynamic and symptomatic effects are almost completely lost. Tolerance develops early, within 24 hours of the initiation of therapy, and it cannot be overcome with the administration of higher doses. This loss of therapeutic effect in the face of continued therapeutic plasma concentrations has led to more than three decades of intense investigation concerning the etiology of tolerance, a controversy that continues to this day. Shortly after the initial description of tolerance, it was recognized that nitrate effects could be maintained using dosing regimens that allowed for a nitrate-free or low-nitrate concentration for several hours each day. This observation led to the adoption of intermittent or eccentric dosing regimens that now represent the standard of care, particularly in the setting of stable exertional angina. The mechanisms of nitrate tolerance have been the subject of extensive investigation for decades. Several hypotheses have been proposed, although agreement concerning a single unifying hypothesis has never been obtained. A brief overview of approaches to this classic pharmacologic question is outlined below.


Biotransformation Hypothesis


The observation that nitrate concentrations remained at therapeutic levels in the setting of tolerance ruled out a pharmacokinetic etiology and suggested that the loss of their effect was secondary to a decrease in their bioactivation. Although the role of nitrates as NO donors was not yet understood, it was known that nitrate effects were based on enzymatic biotransformation. Initial hypothesis concerning the etiology of nitrate tolerance suggested it was secondary to impaired biotransformation during sustained therapy. Classic investigations by Needleman et al demonstrated that nitrate biotransformation was dependent on reduced sulfhydryl groups and that depletion of these as substrate or cofactors led to loss of nitrate effects. This sulfhydryl depletion hypothesis spawned a series of investigations that attempted to prevent or reverse nitrate tolerance using thiol donors such as N-acetylcysteine. Although substantial in vitro data supported this view, the use of thiol donors to prevent nitrate tolerance was never adopted into clinical practice. In general, early investigations of reduced nitrate biotransformation as the basis of nitrate tolerance were inconclusive, hindered by the fact that the enzyme responsible for the biotransformation process had not been identified. In 2002, two decades after Needleman proposed the sulfhydryl depletion hypothesis, the importance of abnormalities of biotransformation in the setting of tolerance was confirmed by Chen and colleagues with their description of mALDH-2 as the responsible enzyme. This finding has greatly improved the understanding of the phenomenon of nitrate tolerance and is closely linked to the free radical hypothesis of tolerance, which is presented in more detail below.


Neurohormonal Hypothesis


In the late 1980s, a number of investigators proposed the neurohormonal hypothesis, which states that the loss of nitrate effects is mediated by reflex activation of the renin-angiotensin and sympathetic nervous systems secondary to the acute effects of nitrates on loading conditions. This hypothesis was supported by observations that nitrate therapy was associated with evidence of neurohormonal activation along with evidence of plasma volume expansion. Further studies suggested that tolerance could be prevented with concurrent use of angiotensin-converting enzyme (ACE) inhibitors or diuretics, although other observations did not confirm these findings. Overall, the neurohormonal hypothesis did not lead to a clinical solution to the problem of nitrate tolerance, but it did remind investigators that powerful vasoactive agents, such as the organic nitrates, could induce counterregulatory responses, probably at multiple levels.


Free Radical Hypothesis


In 1995, Münzel and colleagues reported an observation that had great impact on the field of nitrate pharmacology. They demonstrated that sustained exposure to GTN was associated with increased vascular free radical production and that the endothelium was the source of these free radicals. They also documented that tolerance was reversed by exposure to the antioxidant liposomal superoxide dismutase, which restored responses to GTN and to the endothelium-dependent vasodilator acetylcholine. Based on these findings, the authors proposed the free radical hypothesis of nitrate tolerance, in which a nitrate-induced increase in free radical production limits nitrate responsiveness. The mechanism of the nitrate-induced free radical production has been extensively explored in both animal and human models. Multiple enzymatic sources have been suggested, including membrane-bound nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, and endothelial NO synthase itself. Animal investigations have documented an important role for angiotensin II in this process. Although the inciting cause was not clear, GTN therapy increased angiotensin II production, increasing NADPH production of superoxide anion. The concurrent administration of an angiotensin II receptor inhibitor prevented the increase in superoxide anion production, maintaining nitrate responsiveness. This pathway deserves emphasis because it is known that hydralazine has antioxidant activity that inhibits NADPH-mediated production of superoxide anion and provides a potential explanation for the beneficial effect of hydralazine in combination with isosorbide dinitrate in CHF.


How an increase in vascular free radical production leads to a decrease in nitrate responsiveness is unclear. Possibilities include 1) abnormalities in nitrate biotransformation secondary to the oxidative state, 2) a decrease in net NO bioavailability secondary to free radical quenching of NO, and 3) free radical–induced changes in signal transduction. Discovery and investigation of the central role of mALDH-2 in GTN biotransformation provided a unifying hypothesis concerning the mechanism of nitrate biotransformation and the development of tolerance. This enzyme is essential to the bioactivity of GTN at therapeutic concentrations, but prolonged exposure to GTN is associated with oxidative inhibition of its function. This oxidative inhibition of mALDH-2 activity provides a mechanism linking nitrate-induced free-radical production with inhibition of nitrate biotransformation with tolerance. What is not clear is what triggers the initial increase in reactive oxygen species. Of note, recent evidence suggests that increased superoxide formation may result directly from GTN mALDH-2 biotransformation, although other potential sources have been suggested.


Despite the importance of mALDH-2 as a mechanism of nitrate biotransformation and tolerance, it is clear that other mechanisms are involved. Münzel and colleagues recently summarized the complexity of this area, but a full description is beyond the scope of this chapter. Important highlights include the observation that mALDH-2 does not mediate the biotransformation of isosorbide dinitrate and isosorbide-5-mononitrate. These nitrates, as well as high concentrations of GTN, are biotransformed by other enzyme systems that may include glutathione-S-transferases, xanthine oxidoreductase, and CYP. Furthermore, tolerance develops in response to all nitrates, emphasizing that this phenomenon must also be more than abnormalities in mALDH-2 function. The alternative mechanisms of tolerance remain poorly defined, but it appears that a decrease in NO bioavailability from increased free radical production may be a feature common to all nitrates. Furthermore, the same increase in free radicals may lead to oxidative inhibition of other nitrate biotransformation enzymes, as well as those involved in NO signal transduction, such as guanylate cyclase. Finally, the fact that therapy with GTN causes abnormalities in endothelial NO synthase function is noteworthy, emphasizing the potential of organic nitrates to have quite unexpected biologic effects. In normal volunteers, transdermal GTN causes abnormalities in NO synthase function, markedly inhibiting their responses to the NO synthase inhibitor L-NMMA. This response appears to be secondary to reduction of tetrahydrobiopterin, which is oxidized by GTN-induced increases in peroxynitrite. This in turn leads to the phenomenon of NO synthase uncoupling, with the result that this enzyme yields superoxide anion rather than NO. The resulting increase in superoxide leads to further dysfunction of NO synthase in a positive feedback mechanism.


Nonhemodynamic Effects of Organic Nitrates


A number of studies have documented that the organic nitrates can inhibit platelet aggregation. Platelets produce NO, which acts to inhibit granule release and aggregation. GTN has been shown to inhibit platelet aggregation both in vitro and in a number of human experiments. Whether tolerance develops to this antiplatelet effect is controversial, and in general, the clinical relevance of this effect is unclear; however, it has been postulated that this effect may be particularly important in the setting of acute coronary syndromes (ACS), although the efficacy of GTNs in this situation remains only hypothetical.


Another impact of the organic nitrates unrelated to their hemodynamic effects is their ability to precondition. GTN has been shown to have important preconditioning effects in a number of animal models. In humans, short-term exposure to a GTN leads to decreased evidence of ischemia during percutaneous coronary intervention (PCI) and in the setting of exercise-induced ischemia. Human models have documented that development of the preconditioned phenotype is inhibited by the administration of an antioxidant during exposure to GTN. Little information is available concerning the ability of other organic nitrates to precondition, but one human study found that protection from ischemia and reperfusion was not found after administration of isosorbide-5-mononitrate.


The observation that mALDH-2 plays a critical role in the development of ischemic preconditioning draws an interesting link between the biotransformation of GTN, the subsequent increase in free radical bioavailability, and the preconditioning phenotype. To date, this capacity of the GTN to precondition has not found any meaningful clinical application. Of note, a recent report on normal volunteers suggested that the acute preconditioning effects of a single short-term exposure (2 hours) to transdermal GTN were lost during sustained daily exposure to GTN, suggesting that tolerance develops to the preconditioning effect.


As the importance of NO bioavailability in CV disease became clear, it was believed that NO donors, such as the organic nitrates, would have a beneficial effect as supplemental sources of NO. Although this benefit remained a matter of conjecture, it was never believed that organic nitrates could have adverse effects on vascular function. Given this background, the demonstration that animals exposed to GTN developed significant abnormalities of endothelial function was unexpected. GTN has long been considered a non–endothelium-dependent vasodilator with actions confined to vascular smooth muscle cells. Nevertheless, these observations in animals were followed by human experiments, which confirmed that sustained nitrate exposure causes significant and surprising abnormalities of vascular function. In patients with CAD, 48 hours of intravenous GTN increased the sensitivity of the arterial resistance vessels to angiotensin II and phenylephrine. Further studies revealed that continuous GTN therapy also caused important abnormalities in endothelial function in normal volunteers and worsened endothelial function in those with CAD. Similar abnormalities of vascular function have also been documented during both intermittent transdermal GTN and once-daily administration of isosorbide-5-mononitrate.


The importance of NO biology in the function of the sympathetic nervous system has been recognized. Nitric oxide and NO donors have been shown to have inhibitory effects on sympathetic outflow at multiple sites, both peripheral and central. When given acutely, the pronounced hemodynamic effects of these drugs causes reflex stimulation of the sympathetic nervous system, which complicates experimental observations in this area. Animal observations suggest that sustained nitrate therapy might be associated with an increase in sympathetic outflow. Interestingly, in a human model, continuous transdermal GTN caused a reduction in tonic and reflex modulation of heart rate, leading to a relatively greater sympathetic influence. The overall effect was a blunting of spontaneous baroreflex function, an abnormality usually associated with specific CV diseases. The clinical implications of this are not clear but are an example of a “nitrate effect” associated with abnormalities that generally are believed to have a negative effect on prognosis.


Current Perspectives on Therapy with Organic Nitrates


Despite approximately 150 years of clinical use, many biologic effects of the organic nitrates remain poorly understood. Given the current state of knowledge, it appears that these drugs can have both beneficial and potentially harmful effects, depending on how they are prescribed and for what indication. The understanding of the potential for harm is limited, but it warrants both attention and further study.


In terms of benefits, the effectiveness of organic nitrates in the relief of episodes of angina is unquestioned. Although their use in ACS and acute decompensated heart failure has not been evaluated in large-scale clinical trials, their utility is these settings is widely accepted. They are effective in the treatment of chronic angina, although the development of tolerance is a limitation, and their impact in long-term clinical outcome has never been tested in this population. The clear beneficial role of nitrates in combination with hydralazine in the treatment of chronic CHF was previously discussed. Of note, it has yet to be documented that a nitrate alone can benefit long-term outcome in this patient population. It is now known the GTN has preconditioning effects that limit the adverse effects of ischemia and reperfusion; however, as with other preconditioning approaches, a clearly beneficial application of this biologic effect of nitrates has yet to be defined.


With respect to adverse effects, the finding that sustained nitrate therapy is associated with increased free radical production and evidence of endothelial dysfunction suggests the possibility that these drugs could have harmful effects during chronic therapy. Although these nitrate-induced abnormalities in vascular function have been documented for more than a decade, they have not yet modified the clinical utilization of nitrate therapy. The clinical implications of these findings are unclear, serving to highlight the paucity of clinical data available with respect to clinical outcome during sustained administration with nitrates. Nitrates have been tested in relatively large numbers of patients in the early postinfarction period ; however, the treatment and follow-up periods of these trials were too short to examine the question of safety. Of note, two retrospective analyses of post-MI patients have suggested that long-acting nitrate therapy is associated with an increased mortality rate. Although studies in heart failure (discussed above) have documented a beneficial effect of isosorbide-5-mononitrate when given in combination with hydralazine, the safety and efficacy of a nitrate alone in the setting of chronic heart failure has never been examined. Although it is unclear whether clinical outcome studies will ever be completed, it can be stated that there should be no assumption of clinical safety when these drugs are used as long-term therapy.


It is also important to recall that almost all available information concerning the development of tolerance and/or endothelial dysfunction during nitrate therapy was obtained in normal volunteers or patients not taking other cardiac medications. Of note are both animal and human studies in which tolerance is prevented by concurrent therapy with 3-hydroxy-3-methylglutaryl coenzyme A (HMG Co-A) reductase inhibitors as well as agents that inhibit the renin-angiotensin system. In the absence of concurrent therapy with either of these classes of medications, studies were carried out in patients with stable angina; these represent the classic human model of tolerance to the symptomatic benefit of nitrates. A recent study in humans demonstrated that atorvastatin given concomitantly with continuous GTN completely prevented the development of both tolerance and abnormalities of endothelial dysfunction during sustained therapy. Given this background, it can be said that some historic models of nitrate tolerance should be revisited in the current era of pharmacotherapy.




Calcium Channel Blockers


Calcium channel blockers (CCBs) are agents that inhibit several specific calcium-dependent functions in the cardiovascular system. By decreasing vascular smooth muscle contraction and tone they produce peripheral and coronary vasodilation. The non-DHPs (NDHPs) have a negative inotropic effect, which is an undesired action if it becomes excessive. Certain CCBs (e.g., verapamil, diltiazem) inhibit calcium-dependent sinoatrial (SA) and atrioventricular (AV) nodal conduction. The CCBs are approved for use in hypertension, angina pectoris, and acute supraventricular tachycardias. In the United States, the most commonly used available CCBs are diltiazem, verapamil, nifedipine, amlodipine, and felodipine. Bepridil, isradipine, and nicardipine are available but are used relatively infrequently; nimodipine is usually used only for subarachnoid hemorrhage or ruptured cerebral aneurysm.


Fundamental Mechanisms of Calcium Channel Blockers


Calcium Channel as Site of Action


CCBs interfere with the entry of Ca 2+ into cells through voltage-dependent L- and T-type calcium channels. The major cardiovascular sites of action are 1) vascular smooth muscle cells, 2) cardiac myocytes, and 3) SA and AV nodal cells. By binding to specific sites in the proteins of the calcium channel known as subunits, these agents are able to diminish the degree to which the calcium channel pores open in response to voltage depolarization ( Figure 7-4 ).




FIGURE 7-4


Proposed arrangement of the polypeptide chain of the channel forming a 12 subunit of the L-type calcium channel in humans. Four motifs—I, II, III, and IV—are repetitive, and each consists of six putative transmembrane segments. Both the N terminal and the C terminal point to the cytoplasm. Rings separate the segments encoded by numbered exons. The transmembrane segments encoded by alternative exons 8 or 8A, 21, or 22, and 31 or 32 are shown. Sequences encoded by invariant exons 7, 33, and 45, which are subject to constitutive splicing, are also shown. Exons 40, 41, and 42 are subject to alternative splicing. Putative sites of glycosylation and of phosphorylation involving protein kinase C ( C ) and protein kinase A ( A ) are shown, as are the discrete binding areas of the three types of calcium antagonists—phenylalkylamine (verapamil-like), benzothiazepine (diltiazem-like), and dihydropyridine (nifedipine-like).

(From Abernethy DR, Schwartz JB. Calcium-antagonist drugs. N Engl J Med 1999;341:1448.)


Molecular Structure


The calcium channel consists of four high-molecular-weight subunits: α1, α2, β, and γ. Of these, the α1 subunit contains the calcium channel pores and the binding sites for CCBs. The subunits have a complex structure with four major domains (see Figure 7-4 ), each with six transmembrane units. The calcium channel pores exist between the fifth and sixth units, and the voltage sensor is located near the fourth transmembrane unit of each domain.


Two regulatory aspects of calcium channel blockade are important. First, when cyclic adenosine monophosphate (cAMP) activates protein kinase A to phosphorylate the calcium channel, a number of phosphorylation sites are available on the COOH-terminal portion of each of the α1 subunits. Such phosphorylation allows the channel to persist in a more open state. Second, the β subunit binds to the cytoplasmic link between domains I and II of the γ1 subunit and thereby enhances calcium channel opening.


Drug Binding Sites


At least three binding sites exist for these drugs, commonly identified by the prototype agents verapamil, nifedipine, and diltiazem, respectively; these are known as the V-, or phenylalkylamine-; N-, or DHP-; and D-, or benzothiazepine-, binding sites. The N-binding site is also termed the DHP site, to which all DHPs are thought to bind. Each of the different agents binds to specified sites on various domains, and none binds to all of the pores in all of the domains. Thus, calcium channel blockade can never be complete.


Calcium Channels: L and T Types


The most important property of the CCBs is to selectively inhibit the inward flow of charge-bearing Ca 2+ when the calcium channel becomes permeable, or “open.” At least two types of calcium channels are relevant to the treatment of CV disorders: the L and T types. The major calcium channel related to pharmacologic antagonism, the voltage-gated L-type (long-acting, slowly activating) channel, is blocked by all available CCBs. The function of the L-type channel is to allow entry of sufficient Ca 2+ for initiation of contraction by calcium-induced intracellular calcium release from the sarcoplasmic reticulum.


The T-type (transient) channel appears at more negative potentials than the L type and probably plays an important role in the initial depolarization of SA and AV nodal tissue. The L-type calcium channel is found in vascular smooth muscle, in nonvascular smooth muscle in many tissues, and in a number of noncontractile tissues. Blockade of the L-type channel is responsible for the pharmacologic actions of the available CCBs.


Pharmacologic Properties of Calcium Channel Blockers


Pharmacodynamic Effects


Despite their structural diversity and binding differences, CCBs display many common important pharmacologic actions; however, there are significant differences between the sites of action of the DHPs and NDHPs ( Table 7-3 ).



TABLE 7-3

Vasodilator Potency and Inotropic, Chronotropic, and Dromotropic Effects of Calcium Channel Blockers




















































AMLODIPINE DILTIAZEM NIFEDIPINE VERAPAMIL
Heart rate ↑/–
Sinoatrial node conduction
Atrioventricular node conduction
Myocardial contractility ↓/– ↓/– ↓↓
Neurohormonal activation ↑/–
Vascular dilation ↑↑ ↑↑
Coronary flow

↓, decrease; –, no change; ↑, increase

Modified from Abernethy DR. Pharmacologic and pharmacokinetic profile of mibefradil, a T- and L-type calcium channel antagonist. Am J Cardiol 1997;80:4c-11c.


Major Cardiovascular Actions of Calcium Channel Blockers




  • 1

    Vasodilation is more marked in arterial and arteriolar vessels than on veins and includes the coronary vasculature; veins do not appreciably dilate with CCBs.


  • 2

    Negative chronotropic and dromotropic effects are seen on the SA and AV nodal conducting tissue (NDHP agents only).


  • 3

    Negative inotropic effects are seen on myocardial cells; in the case of DHPs, this effect may be offset by reflex adrenergic stimulation after peripheral vasodilation.



Classification of Calcium Channel Blockers


The differing pharmacodynamic effects of various CCBs accounts for their classification. All the DHPs bind to the same sites on the α1 subunit and exert a greater Ca 2+ inhibitory effect on vascular smooth muscle than on the myocardium, which explains their common property of vascular selectivity; thus their major hemodynamic and therapeutic effect is peripheral and coronary vasodilation.


Nifedipine is the prototypical DHP. The fast-acting capsular form produces rapid vasodilation, alleviates hypertension, and terminates attacks of coronary spasm. However, the brisk peripheral vasodilation produced by this formulation may result in significant hypotension and reflex adrenergic activation that often causes tachycardia and stimulation of the sympathetic and renin-angiotensin systems. The introduction of truly long-acting DHP compounds—such as amlodipine or sustained-release formulations of nifedipine, felodipine, or isradipine—has resulted in substantially fewer symptoms from the vasodilatory side effects. It is a commonly held belief that the short-acting DHPs, particularly nifedipine, account for the majority of presumed negative or adverse clinical results in many older trials. The second-generation DHPs are distinguished by a longer half-life, as in the case of amlodipine, or by a greater vascular selectivity.


Although each binds to a different site on the α1 subunit, the NDHPs verapamil and diltiazem have many properties in common. Both act on nodal (SA and AV) tissue and are therapeutically effective in supraventricular tachycardias. Both decrease the sinus discharge rate. These drugs inhibit myocardial contractility more than the DHPs; in effect, they are less vascularly selective. Both verapamil and diltiazem have greater effects on the AV node than on the SA node, and the explanation for this may relate to frequency dependence; thus there is better access to the binding sites when the calcium channel pore is open. During supraventricular tachycardia, the calcium channel of the AV node opens more frequently, so the CCB binds more avidly, hence it more specifically inhibits the AV node to interrupt the reentry circuit.


Regarding side effects, because NDHPs are less active on vascular smooth muscle, they produce fewer vasodilatory adverse reactions than the DHPs. Sinus tachycardia is uncommon, in part because of the inhibitory effects on the SA node. High-degree AV block is a risk with preexisting AV nodal disease or during cotherapy with other AV node–depressant drugs, such as β-blockers. NDHPs have a more marked depressive effect on ventricular function than DHPs. In addition, constipation occurs as a side effect with verapamil but seldom with diltiazem, although the latter may cause peripheral edema.


Vascular Selectivity


The cellular mechanism of vascular smooth muscle contraction differs from that of the myocardium. Although smooth muscle contraction is ultimately calcium dependent, it is the myosin light-chain kinase that is activated by calcium calmodulin. In the human myocardium, Godfraind and associates proposed that the ratios of vasodilation to negative inotropy for the prototype CCBs were 10 : 1 for nifedipine, 1 : 1 for diltiazem, and 1 : 1 for verapamil. Other DHP compounds have even greater vascular selectivity, up to 1000 : 1. In terms of clinical use, these observations provide the basis for considering a clinical division of the CCBs into two groups: DHPs, which include nifedipine and its analogs, and NDHPs, such as verapamil, diltiazem, and their derivatives.


Noncardiovascular Effects


Although highly active on vascular smooth muscle, CCBs have little or no effect on other smooth muscle throughout the body, such as that of the bronchi, gut, or genitourinary tract. These agents may relax uterine smooth muscle and have been used in therapy for preterm contractions, although it is generally recommended that they be stopped before delivery. This action probably reflects variations between tissues in either the structure or function of their calcium channels. Also crucial to the therapeutic applicability of CCBs is the fact that skeletal muscle does not respond to conventional CCBs. As a result, skeletal muscle weakness is not a side effect of calcium channel blockade. In skeletal muscle, depolarization-activated calcium release from the sarcoplasmic reticulum is the principal source of the myoplasmic calcium rise. Thus only the myocardium, not skeletal muscle, responds to calcium entry through the voltage-dependent calcium channels; and the myocardium, not skeletal muscle, has its rise in contractile calcium inhibited by CCBs.


Pharmacokinetics


From the point of view of drug interactions, all of the CCBs are metabolized in the liver by an enzyme system that is inhibited by cimetidine, azole antifungals, and hepatic dysfunction; CCBs are increased in activity by phenytoin and phenobarbital.


Major Indications for Calcium Channel Blockers


Systemic Hypertension


The various CCBs act on peripheral arterioles. They are effective antihypertensive agents in all ethnicities and age groups. All DHPs decrease peripheral vascular resistance and appear to have an additional ill-understood diuretic effect. Verapamil and diltiazem are less powerful vasodilators, and some believe that their negative inotropic effect may contribute to their antihypertensive mechanism. Table 7-4 lists some major hypertension trials that used CCBs.



TABLE 7-4

Selected Characteristics of Trials of Calcium Channel Blockers





































































STUDY AGENTS USED PATIENTS ENROLLED PATIENT CHARACTERISTICS
ALLHAT Amlodipine vs. chlorthalidone vs. lisinopril 33,357 Hypertension and one risk factor for coronary artery disease
INVEST Verapamil slow-release ± trandolapril ± hydrochlorothiazide vs. atenolol ± hydrochlorothiazide ± trandolapril 22,576 Hypertension and coronary artery disease
CONVINCE Extended-release verapamil vs. atenolol or hydrochlorothiazide 16,602 Hypertension and one risk factor for coronary artery disease
NORDIL Diltiazem vs. diuretics + β-blockers 10,881 Hypertension
STOP-2 Felodipine or isradipine vs. conventional antihypertensive agents 6614 Hypertension
INSIGHT Nifedipine gastrointestinal therapeutic system vs. hydrochlorothiazide + amiloride 6321 Hypertension and one risk factor for coronary artery disease
VHAS Verapamil vs. chlorothiazide 1414 Hypertension
MIDAS Isradipine vs. hydrochlorothiazide 883 Hypertension
ABCD Nisoldipine vs. enalapril 470 Hypertension and diabetes mellitus
NICS-EH Nicardipine vs. trichloromethiazide 429 Hypertension and age >60 years
FACET Amlodipine vs. fosinopril 380 Hypertension and diabetes mellitus
CASTEL Nifedipine vs. clonidine or atenolol + chlorthalidone 351 Hypertension and age >65 years

ABCD, Appropriate Blood Pressure Control in Diabetes; ALLHAT, Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack trial; CASTEL, Cardiovascular Study in the Elderly; CONVINCE, Controlled Onset Verapamil Investigation of Cardiovascular Endpoints; FACET, Fosinopril versus Amlodipine Cardiovascular Events Trial; INSIGHT, International Nifedipine Gastrointestinal Therapeutic System Study–Intervention as a Goal in Hypertension Treatment; INVEST, International Verapamil Slow-Release/Trandolapril Study; MIDAS, Multicenter Isradipine Diuretic Atherosclerosis Study; NICS-EH, National Intervention Cooperative Study in Elderly Hypertensives; NORDIL, Nordic Diltiazem; STOP, Swedish Trial in Old Patients with Hypertension; VHAS, Verapamil in Hypertension and Atherosclerosis Study.

Modified from Eisenberg MJ, Brox A, Bestauwros AN. Calcium channel blockers: an update. Am J Med 2004;116:35-43.


Angina Pectoris


Although the antianginal mechanisms of the different types of CCBs differ somewhat, these drugs share some properties, including 1) coronary vasodilation, especially in relation to exercise-induced coronary constriction, and 2) afterload reduction as a result of decreased blood pressure. In the case of verapamil and diltiazem, it is possible that slowing of the sinus node, with a decrease in nonmaximal exercise heart rate and the negative inotropic effect, may contribute to decreased myocardial work.


As coronary dilators, the CCBs have a site of action on the coronary tree different from that of the nitrates. The CCBs act more specifically on the smaller coronary resistance vessels, where the tone is higher, and the calcium inhibitory effect is more marked. CCBs are particularly effective in those types of angina caused by or exacerbated by coronary spasm or constriction, such as Prinzmetal angina or cold-induced angina. An overview of a large number of angina drug trials concluded that the CCBs have a very similar clinical efficacy to β-blockers.


Supraventricular Tachycardia


Through their inhibitory effect on the AV node, verapamil and diltiazem interrupt the reentry circuit in supraventricular tachycardias and are useful in terminating those arrhythmias. They are also effective in slowing the ventricular response in atrial fibrillation (AF) and may be used in chronic AF; the DHPs are ineffective for these arrhythmias because of minimal effects on the SA and AV nodes.


Postinfarct Protection


Verapamil is licensed in Scandinavian countries for postinfarct protection for patients in whom β-blockers are contraindicated. In the Danish Verapamil Infarction Trials (DAVIT-1 and DAVIT-2), a modest protective benefit against death and cardiac ischemic events in post-MI subjects was documented in subjects without a history of heart failure. Diltiazem has been shown to be beneficial in post-MI subjects with relatively normal left ventricular (LV) function and no heart failure. A short-term (2-week) study in non–Q-wave MI patients with high-dose diltiazem reduced the rates of recurrent ischemia and infarction.


Specific Calcium Channel Blockers


Verapamil


After peripheral vasodilation induced by verapamil, the cardiac output and LV ejection fraction do not increase as much as they do with the DHPs, probably owing to the negative inotropic effect and depression of contractility of verapamil.


Pharmacokinetics


The elimination half-life of standard verapamil tablets is usually 3 to 7 hours, but it increases significantly during long-term administration and in patients with liver or renal insufficiency. In significant hepatic dysfunction, the dose of verapamil should be decreased by 50% to 75%. In significant renal dysfunction, such as a creatinine clearance of less than 30 mL/min, the dose should be reduced by 50%. Bioavailability is only 10% to 20% (high first-pass liver metabolism). The parent compound and the active hepatic metabolite, norverapamil, are excreted 75% by the kidneys and 25% by the gastrointestinal (GI) tract. Verapamil is 87% to 93% protein bound.


Dose


Oral Preparations


The usual dosage of the standard preparation is 80 to 120 mg three times daily. During long-term oral dosing, less frequent daily doses are needed (norverapamil metabolites). Slow-release preparations (240 to 480 mg/day) are available and are the usual regimen.


Intravenous Use


For supraventricular reentry tachycardias, a bolus of 5 to 10 mg (0.1 to 0.15 mg/kg) can be administered over 2 minutes and repeated 15 to 20 minutes later if needed. After successful administration, the dose may be stopped or continued at 0.005 mg/kg/min for approximately 30 to 60 minutes, decreasing thereafter. When used for control of the ventricular rate in AF, verapamil may be administered at 0.005 mg/kg/min, increasing as needed, or as an IV bolus of 5 mg, followed by a second bolus of 10 mg if needed. In the presence of myocardial disease or interacting drugs, a very low dosage (0.0001 mg/kg/min) may be infused and titrated upward against the ventricular response. However, safer AV-slowing agents, such as digoxin and adenosine, are available for patients with impaired LV systolic function.


Side Effects


Side effects include headaches, facial flushing, dizziness, and ankle edema—all lower in frequency than with DHPs. Constipation occurs in up to one third of patients who receive verapamil, and the negative inotropic effect of verapamil may precipitate or exacerbate CHF. When IV verapamil is used, the risk of hypotension is increased if the patient is receiving β-blockers or other vasodilators or has depressed cardiac function.


Contraindications


Sick sinus syndrome and preexisting AV nodal disease are relative contraindications to IV and oral verapamil. The effective use of oral verapamil preparations in these conditions may require a pacemaker. In Wolff-Parkinson-White syndrome with AF, IV verapamil may promote antegrade conduction of impulses down the bypass tract, with a risk of very rapid AF and even ventricular fibrillation. In a wide–QRS complex ventricular tachycardia, verapamil is contraindicated because the combined negative inotropic and peripheral vasodilatory effects can be fatal. Furthermore, verapamil is unlikely to terminate a ventricular arrhythmia and should not be used in the setting of moderate or severe LV dysfunction or severe hypotension.


Pregnancy


Category C specifies use only if potential benefit justifies the potential risk to fetus; no well-controlled trials are available.


Diltiazem


Diltiazem is used for the same spectrum of CV disease as verapamil: hypertension, angina pectoris, prevention of AV nodal reentry, tachycardia, and rate control in acute and chronic AF. The side-effect profile is similar, except that constipation is much less common.


Pharmacokinetics


More than 90% of oral diltiazem is absorbed, with approximately 45% bioavailability (first-pass hepatic metabolism). The onset of action is within 15 to 30 minutes, and peak effects occur at 1 to 2 hours. The elimination half-life is 4 to 7 hours, and protein binding is 80% to 90%. Diltiazem is acetylated in the liver to the active metabolite desacetyl diltiazem (40% of the activity of the parent compound), which accumulates during long-term therapy. Only 35% of diltiazem is excreted by the kidneys; the rest is excreted by the GI tract.


Dose


The standard oral dose of short-acting diltiazem is 120 to 360 mg daily in three or four divided daily doses. The slow-release preparations are administered once or twice daily. IV diltiazem (approved for arrhythmias) is administered as 0.25 mg/kg over 2 minutes with electrocardiographic (ECG) and blood pressure monitoring; if the response is inadequate, the dose is repeated as 0.35 mg/kg in 15 to 20 minutes. Acute loading therapy may be followed by an infusion of 5 to 15 mg/h.


Side Effects


Side effects are few and are limited to headaches, dizziness, and ankle edema in 6% to 10% of patients. The extended or slow-release preparations appear to have a side-effect profile similar to that of placebo. Sinus bradycardia and first-degree or higher AV nodal block may be produced by diltiazem. It is important to avoid or reduce dosing in subjects with SA or AV nodal disease. In heart failure with significant LV dysfunction (e.g., ejection fraction <35%), this drug can be hazardous. Exfoliative dermatitis and skin rash occasionally occur, and the side effects of IV diltiazem resemble those of IV verapamil.


Contraindications


Contraindications are similar to those of verapamil: preexisting depression of the SA or AV node, hypotension, low ejection fraction, heart failure, and AF associated with Wolff-Parkinson-White syndrome. LV failure ejection fraction of less than 40% after MI is a clear contraindication.


Pregnancy


Category C specifies use only if potential benefit justifies the potential risk to the fetus; no well-controlled trials are available.


Dihydropyridines


The major therapeutic action of the DHPs is arterial and arteriolar dilation, which is responsible for their efficacy in hypertension and angina pectoris as well as in Prinzmetal or variant angina and Raynaud phenomenon. Direct negative inotropic effects of DHPs are minimal. Amlodipine is the CCB of choice in patients with severely depressed LV function, because it does not decrease LV contractility at standard doses. No clinically significant evidence is available showing the effect of DHP on either the SA or the AV node; these agents are not effective in supraventricular arrhythmias; however, they may be more readily combined with β-blockers in hypertension or angina pectoris than the rate-slowing CCBs, with less concern about depression of the SA and AV nodes.


First-Generation Dihydropyridines


Oral nifedipine is the prototypical DHP. It is rapidly absorbed with peak blood levels in 20 to 45 minutes and a duration of action of 4 to 8 hours. Because of its short half-life and difficulty controlling the degree of blood pressure lowering, it is rarely used in its short-acting form. Slow-release forms are currently available and are preferred by most physicians. The dose for the slow-release form is 30 to 90 mg once a day.


Contraindications and Cautions


The short-acting forms are generally contraindicated because of their rapid hypotensive effect in some patients.


Side Effects


Because DHPs have no SA or AV effects, reflex tachycardia may occur if excessive blood pressure lowering occurs. Headache can occur with any of the CCBs, but they occur more frequently with the first-generation DHPs.


Pregnancy


Category C specifies use only if potential benefit justifies the potential risk to the fetus; no well-controlled trials are available.


Second-Generation Calcium Channel Blockers


Theoretically, the more vascular-selective DHPs—such as felodipine, isradipine, amlodipine, and nicardipine—should be safer than nifedipine in the management of angina or hypertension, particularly when LV function is impaired. These drugs may produce adverse effects in patients with CHF, although felodipine and amlodipine appear to be quite safe in patients with depressed LV function. In fact, amlodipine has been shown to have no adverse effect and no benefit compared with placebo in the Prospective Randomized Amlodipine Survival Evaluation (PRAISE) and PRAISE-2 heart failure trials. These compounds are the DHPs of choice in subjects with decreased LV function or a history of heart failure, and they are also popular because of their once-daily dosing schedule.


Although amlodipine is no more vascularly selective than nifedipine, it has unusual pharmacokinetics, including slow onset and offset of binding to the calcium channel site and a prolonged elimination half-life. Based on these pharmacokinetic characteristics and new extensive experience with this agent in both angina and antihypertensive studies, amlodipine has become the DHP of choice for most physicians in the Western Hemisphere.


Drug Interactions of Calcium Channel Blockers


β-Blockers


Verapamil and diltiazem contribute to SA or AV nodal and myocardial depression; in addition, they may interact via hepatic mechanisms with β-blockers metabolized by the liver, such as propranolol and metoprolol. Although these drugs have been successfully combined with β-blockade in the therapy of angina and hypertension, clinicians should monitor patients for possible serious adverse effects when a rate-slowing CCB is combined with a β-blocker.


Digoxin


Verapamil increases blood digoxin levels by decreasing the renal excretion of digoxin. Enhancement of AV nodal block can be serious and even fatal when IV verapamil is administered to patients with digitalis intoxication.


Diltiazem


In general, drug interactions with diltiazem are similar to those of verapamil, but diltiazem has a slight or negligible effect on blood digoxin levels. Although it may be cautiously combined with β-blockade, the combination appears to be no more effective in some studies than high-dose diltiazem alone. Cimetidine may increase diltiazem bioavailability and result in a 50% to 60% increase in plasma diltiazem levels.


Dihydropyridines


The combination of DHPs with β-blockers is safer than that with NDHP CCBs. When LV depression is present, the added negative inotropic effects of a β-blocker and DHP may precipitate overt heart failure, but this is unusual; amlodipine or felodipine is the CCB of choice in such individuals.


Calcium Channel Blockers: The “Safety” Controversy


Beginning in 1995, a question about the safety of all CCBs was raised when a retrospective analysis of the short-acting form of nifedipine appeared to increase heart attacks in ACS patients, but the data were grouped with all CCBs. As prospective trials did not confirm these fears, and as physicians gained experience with the slow-release NDHPs and long-acting DHPs such as amlodipine, this issue gradually died. In fact, several antihypertensive trials have established the safety and benefit of these agents.


There has been a burst of data on the use of CCBs in the past decade, which offers important and reassuring safety data regarding CCB use in patients with ischemic heart disease. The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) was a study of 33,357 patients aged older than 55 years with hypertension and at least one other common heart disease risk factor. The patients were randomized to one of four antihypertensive regimens: chlorthalidone, a diuretic; doxasin, an α-blocker; amlodipine, a CCB; and lisinopril, an ACE inhibitor. The doxasin arm was terminated early because of an increased CV risk; however, no differences were evident in the diuretic, CCB, or ACE-inhibitor arms in terms of the primary outcome of combined fatal CHD and nonfatal MI or all-cause mortality.


A second trial, the Valsartan Antihypertensive Long-term Use Evaluating (VALUE) trial, was designed specifically to test the hypothesis that the angiotensin receptor blocker valsartan would be superior to amlodipine for the same blood pressure (BP) control. A total of 15,245 patients older than 50 years were followed for a mean of 4.2 years, until 1450 events had accumulated. BP was reduced by both treatments, but the amlodipine-based therapies were more effective in BP control, particularly early in the study, achieving 4.0/2.1 mm Hg lower pressure compared with the valsartan group at 1 month, and achieving 1.5/1.3 mm Hg lower pressure at 1 year. Most importantly, no evidence of harm was found in the patient population, and a nonstatistically significant slightly lower overall event rate occurred in the amlodipine group: 810 patients in the valsartan group (25.5 per 1000 patient-years) and 789 in the amlodipine group (24.7 per 1000 patient-years). In addition, of the secondary outcomes, MI occurred more frequently ( P = .02) in the valsartan group compared with the amlodipine group ( Figure 7-5 ). The authors hypothesized that the lower BPs in the calcium channel group may explain the lack of superiority of the angiotensin receptor blocker, but it seems unlikely that it could also explain the statistically significantly lower infarction rate.




FIGURE 7-5


Systolic and diastolic blood pressure ( BP ) and differences (valsartan and amlodipine) in BP between treatment groups during follow-up. BP difference between the two groups in the Valsartan Antihypertensive Long-Term Use Evaluating (VALUE) trial was significant ( P < .0001) at every time point, favoring the amlodipine-based regimen. Overall differences in systolic BP were 2.23 mm Hg (standard error, 0.18); overall differences in diastolic BP were 1.59 mm Hg (standard error, 0.11).

(Modified from Julius S, Kjeldsen V, Weber M, et al, for the VALUE trial group. Outcomes in hypertensive patients at high cardiovascular risk treated with regimens based on valsartan or amlodipine: the VALUE randomised trial. Lancet 2004;363:2024.)


The Comparison of Amlodipine Versus Enlapril to Limit Occurrences of Thrombosis (CAMELOT) trial randomized 1992 patients with angiographically proven CAD who were normotensive at baseline (mean BP, 129/78 mm Hg for both arms) to amlodipine or enalapril versus placebo. The incidence of CV events was 23% in the placebo arm, 20% with enalapril, and 17% with amlodipine, with similar BP results in the two active treatment arms that were significantly lower than the placebo; this reduction was driven solely by coronary revascularization and angina, not hard endpoints, although a nonsignificant trend was observed toward benefit in hard CV endpoints for both treatment arms compared with placebo. Importantly, given data from Action to Control Cardiovascular Risk in Diabetes (ACCORD)—which suggests that aggressive therapy in high-risk diabetic patients to a systolic BP less than 120 mm Hg may not be beneficial compared with a goal of less than 140 mm Hg —the mean systolic BP after therapy dropped by only 4.5 mm Hg, meaning that BPs in the treatment arm were still higher than the goal in the ACCORD trial.


As is commonly found in clinical practice, combination therapy to reach target BP is often required, which limits the generalizability of prior trials that tested single-agent regimens. More recent trials have attempted to address this concern by comparing predefined combination therapies. One of these trials to assess the prevention of CV events with an antihypertensive regimen of amlodipine, adding perindopril as required, versus atenolol, adding bendroflumethiazide as required, was the Anglo-Scandinavian Cardiac Outcomes Trial–Blood Pressure Lowering Arm (ASCOT-BPLA). It examined of the efficacy of CCBs or β-blockers as first-line therapy, in combination with a renin-angiotensin-aldosterone system (RAAS) inhibitor, versus diuretic in 19,257 patients with hypertension who were 40 to 79 years old and at high CV risk. The primary outcome was a combined endpoint of fatal coronary events and nonfatal MI. In the CCB arm, 39% of patients were concomitantly on an RAAS inhibitor, and in the β-blocker arm, 49% were concomitantly on a diuretic. The trial was stopped early because of increased CV events in the β-blocker arm. Patients in the CCB arm had fewer CV events (27.4 vs. 32.8 per 1000 patient-years; relative risk [RR], 0.84), a lower all-cause mortality (13.9% vs. 15.5%; RR, 0.89), and although not a primary endpoint, less incidence of diabetes, the latter reflecting known side effects of both β-blocker and thiazide diuretics. In contrast to the beneficial finding of NDHP in the ASCOT trial, another large trial of DHP CCBs did not demonstrate any benefit compared with RAAS inhibition. The International Verapamil-Trandolapril Study (INVEST) randomized 22,576 patients with CAD to verapamil or trandolarpril and found no difference in either arm in terms of the primary endpoint of first occurrence of all-cause death, nonfatal MI, or nonfatal stroke.


An important question addressed partially in ASCOT is what to do when multiple agents need to be started in patients at high CV risk. In particular, it is uncertain which agent to add to RAAS inhibition, which is often already indicated in patients with diabetes, in post-MI patients, and in those with established CV disease. The largest trial to address this question is the Avoiding Cardiovascular Events through Combination Therapy in Patients Living with Systolic Hypertension (ACCOMPLISH) trial, which randomized 11,506 patients to a two-drug regimen. Both arms included benazepril, and the trial compared the addition of the CCB amlodipine versus diuretic therapy with hydrochlorthiazide. The primary outcome of the trial was the composite of death from CV causes, nonfatal MI, nonfatal stroke, hospitalization for angina, resuscitation after sudden cardiac arrest, and coronary revascularization. BP goals were achieved in both arms, yet the combination with the CCB had a 2.2% absolute risk reduction in the primary endpoint compared with the diuretic regimen (hazard ratio [HR], 0.80; 95% confidence interval [CI], 0.72 to 0.90; P < .001; Figure 7-6 ).




FIGURE 7-6


Cardiovascular morbidity and mortality with combination therapy. Primary outcomes in the Avoiding Cardiovascular Events through Combination Therapy in Patients Living with Systolic Hypertension (ACCOMPLISH) trial were significantly different and favored the use of benazepril-amlodipine to amlodipine-hydrochlorothiazide. The relative risk reduction was 20% (hazard ratio, 0.80; 95% confidence interval, 0.72 to 0.90; P < .001).

(From Jamerson K, Weber MA, Bakris GL, et al, for the ACCOMPLISH trial investigators. Benazepril plus amlodipine or hydrochlorothiazide for hypertension in high-risk patients. N Engl J Med 2008;359[23]:2417-2428.)


There is a theoretical concern that DHP CCBs may worsen renal function, in particular in patients with diabetes. In contrast to the effect of RAAS inhibitors, CCB may preferentially vasodilate the afferent arteriole and thereby worsen renal function. This was specifically evaluated in the Gauging Albuminuria Reduction with Lotrel in Diabetic Patients with Hypertension (GUARD) trial, which randomized 332 patients with hypertension and albuminuric type 2 diabetes to benazepril, with either hydrochlorthiazide or amlodipine, and followed them for 1 year. This was a noninferiority trial, and the primary endpoint was urinary creatinine ratio as a measure of albuminuria. Both arms demonstrated a regression in proteinuria that likely reflected improved BP control. However, a near 30% change favored the combination of RAAS inhibitor and diuretic, although these findings should be tempered by the more rapid decline in estimated glomerular filtration rate (GFR) in the diuretic arm, which may have biased the results. The results of the GUARD and ACCOMPLISH trials together make tailoring combined therapy more challenging for the physician, in that GUARD suggests that a trade-off may exist such that reducing CV risk may come at the cost of proteinuria when using a combined DHP and CCB. Given the small size of the trial and the confounding progression in kidney disease, this issue requires further investigation.


Regarding the particular issue of whether to use a DHP instead of an NDHP CCB for the treatment of essential hypertension, concern has been raised over the use of a short-acting DHP (nifedipine) and NDHP (verapamil, diltiazem) in patients at high risk for MI. This concern came from an observational study conducted in the 1990s. In this case-control study, patients treated with nifedipine, diltiazem, or verapamil had an increased risk for MI that was not seen with other antihypertensives, particularly when these drugs were used at high doses. However, this finding was likely confounded by the fact that the use of calcium channel blockade is much higher in patients with symptomatic CAD given the potent role these agents have as antianginal medications. It is important to note that no concern has been reported for the long-acting DHP CCBs and any increased risk of MI in patients treated for essential hypertension in multiple trials. The INVEST investigators specifically examined the risk of CV outcomes between long-acting verapamil versus atenolol in a subgroup of patients with prior MI and found no difference between treatment arms. Although results of substudies must be interpreted with caution, these data in the context of other studies suggest that NDHPs are at least as effective as β-blocker therapy.


In summary, based on the trials above, convincing evidence suggests that DHP CCBs are safe and efficacious for most patients with high CV risk who require further BP control or angina treatment, and that in particular, these are useful in combination therapy. Concerns initially raised about the risks observed in studies of short-acting CCBs are not seen in contemporary trials of long-acting CCBs. In a meta-analysis that included 27 trials and 175,634 patients, the authors examined DHP CCBs versus others agents or placebo in patients with hypertension who also had other high-risk features and found that although the odds ratio (OR) for a heart failure was increased (and may reflect the side effect of pedal edema), the OR was 0.96 ( P = .026) for all-cause death favoring DHP CCBs, with no increase in MI in the subgroup of patients with CAD.




β-Adrenergic Blockers


As a major pharmacotherapeutic advance, β-blockers were initially conceived for the treatment of patients with angina pectoris and arrhythmias; however, they also have therapeutic effects in many other clinical disorders, including systemic hypertension, hypertrophic cardiomyopathy, congestive cardiomyopathy, mitral valve prolapse, aortic dissection, silent myocardial ischemia, migraine, glaucoma, essential tremor, and thyrotoxicosis. β-Blockers have been effective in treating unstable angina and in reducing the risks of CV death and nonfatal reinfarction in patients who have survived an acute MI. β-Adrenergic receptor blockade is also a potential treatment modality, with and without fibrinolytic therapy, to reduce the extent of myocardial injury and death during the hyperacute phase of MI.


β-Adrenergic Receptors


The effects of an endogenous hormone or exogenous drug ultimately depend on physiochemical interactions with macromolecular structures of cells called receptors. Agonists interact with a receptor and elicit a response; antagonists interact with receptors and prevent the action of agonists.


In the case of catecholamine action, the circulating hormone or drug (“first messenger”) interacts with its specific receptor on the external surface of the target cells. The drug hormone/receptor complex, mediated by the G protein (Gs), activates the adenyl cyclase enzyme on the internal surface of the plasma membrane of the target cell, which accelerates the intracellular formation of cAMP, whereupon cAMP-dependent protein kinases (“second messengers”) then stimulate or inhibit various metabolic or physiologic processes. Catecholamine-induced increases in intracellular cAMP are usually associated with the stimulation of β-adrenergic receptors, whereas α-adrenergic receptor stimulation is mediated by glycoprotein Gi and is associated with lower concentrations of cAMP and possibly increased amounts of GMP in the cell. These different receptor effects may result in the production of opposing physiologic actions from catecholamines, depending on which adrenergic receptor system is activated.


Most research on receptor action previously bypassed the initial binding step and the intermediate steps and examined either the accumulation of cAMP or the end step, the physiologic effect. Radioactive agonists or antagonists (radioligands) that attach to and label the receptors have been used to study binding and hormone action. The cloning of adrenergic receptors has also revealed important clues about receptor function. The crystal structure of the human β-adrenergic receptor has also been identified.


In contrast to the older concept of adrenergic receptors as static entities in cells that simply initiate the chain of events, newer theories hold that the adrenergic receptors are subject to a wide variety of controlling influences that result in dynamic regulation of adrenergic receptor sites or their sensitivity to catecholamines or both. Changes in tissue concentration of receptor sites are probably involved in mediating important fluctuations in tissue sensitivity to drug action. These principles may have significant clinical and therapeutic implications. For example, an apparent increase in the number of β-adrenergic receptors, and thus a supersensitivity to agonists, may be induced by long-term exposure to antagonists. With prolonged adrenergic receptor blocker therapy, receptor occupancy by catecholamines can be diminished, and the number of available receptors can be increased. When the β-adrenergic receptor blocker is suddenly withdrawn, an increased pool of sensitive receptors is available for endogenous catecholamine stimulation. The resultant adrenergic stimulation may precipitate unstable angina pectoris, an MI, or both. Specific gene polymorphisms of both the β1 and β2 receptors may also influence the pharmacologic response to β-blocking agents.


Effects in Angina Pectoris


Ahlquist demonstrated that sympathetic innervation of the heart causes the release of norepinephrine, activating β-adrenergic receptors in myocardial cells. This adrenergic stimulation causes an increment in heart rate, isometric contractile force, and maximal velocity of muscle-fiber shortening, all of which lead to an increase in cardiac work and myocardial oxygen consumption. The decrease in intraventricular pressure and volume caused by the sympathetic-mediated enhancement of cardiac contractility tends to reduce myocardial oxygen consumption by reducing myocardial wall tension (LaPlace’s law). Although there is a net increase in myocardial oxygen demand, this is normally balanced by an increase in coronary blood flow. Angina pectoris is believed to occur when oxygen demand exceeds supply (i.e., when coronary blood flow is restricted by coronary atherosclerosis). Because the conditions that precipitate anginal attacks—exercise, emotional stress, food—cause an increase in sympathetic cardiac activity, it might be expected that blockade of cardiac β-adrenergic receptors would relieve anginal symptoms. It is on this basis that the early clinical studies with β-blocking drugs in patients with angina pectoris were initiated.


Three main factors contribute to the myocardial oxygen requirements of the LV: heart rate, ventricular systolic pressure , and size of the LV . Of these, heart rate and systolic pressure appear to be important, and the product of heart rate multiplied by the systolic BP is a reliable index for predicting the precipitation of angina in a given patient ; however, myocardial contractility may be even more important.


The reduction in heart rate effected by β-blockade has two favorable consequences: a decrease in blood pressure, thus reducing myocardial oxygen needs, and a longer diastolic filling time associated with a slower heart rate, allowing for increased coronary perfusion. β-Blockade also reduces exercise-induced BP increments, the velocity of cardiac contraction, and oxygen consumption at any patient’s workload ( Box 7-1 ). After treatment, a reduced heart rate variability, a marker for abnormal autonomic control of the heart, or low exercise tolerance may predict those patients who will respond best to treatment with β-blockade. Despite the favorable effects on heart rate, the blunting of myocardial contractility with β-blockers may be the primary mechanism of their antianginal benefit. In normal human coronary arteries, β2-adrenergic receptor–mediated vasodilation enhances coronary perfusion, an effect that is impaired by severe atherosclerosis.



Box 7-1

Possible Mechanisms by Which β-Adrenergic–Blocking Agents Protect the Ischemic Myocardium





  • Reduction in myocardial consumption, heart rate, blood pressure, and myocardial contractility



  • Augmentation of coronary blood flow, increase in diastolic perfusion time by heart rate reduction, augmentation of collateral blood flow, and redistribution of blood flow to ischemic areas



  • Prevention or attenuation of atherosclerotic plaque rupture and subsequent coronary thrombosis



  • Alterations in myocardial substrate utilization



  • Decrease in microvascular damage



  • Stabilization of cell and lysosomal membranes



  • Shift of oxyhemoglobin dissociation curve to the right



  • Inhibition of platelet aggregation



  • Inhibition of myocardial apoptosis, which allows natural cell regeneration to occur



From Frishman WH. Alpha- and beta-adrenergic blocking drugs. In Frishman WH, Sonnenblick EH, Sica DA, editors: Cardiovascular pharmacotherapeutics, 2nd ed. New York, 2003, McGraw-Hill, pp 67-97.


Studies in dogs have shown that propranolol causes a decrease in coronary blood flow. However, subsequent experimental animal studies have demonstrated that β-blocker–induced shunting occurs in the coronary circulation, maintaining blood flow to ischemic areas, especially in the subendocardial region. In humans, concomitant with the decrease in myocardial oxygen consumption, β-blockers can cause a reduction in coronary blood flow and an increase in coronary vascular resistance. On the basis of coronary autoregulation, the overall reduction in myocardial oxygen needs with β-blockers may be sufficient cause for this clinically tolerated decrease in coronary blood flow.


Virtually all β-blockers produce some degree of increased work capacity without pain in patients with angina pectoris, regardless of whether they have partial agonist activity, α-adrenergic receptor-blocking effects, direct vasodilating effects, membrane-stabilizing activity, or general or selective β-blocking properties. Therefore, it must be concluded that this results from their common property: blockade of cardiac β-adrenergic receptors ( Table 7-5 ). Both d- and l-propranolol have membrane-stabilizing activity, but only l-propranolol has significant β-blocking activity. The racemic mixture (d,l-propranolol) causes decreases in both heart rate and force of contraction in dogs, whereas the d-isomer has hardly any β-adrenergic receptor–blocking effect. In humans, d-propranolol, which has “membrane” activity but no β-blocking properties, has been found to be ineffective in relieving angina pectoris even at very high doses.



TABLE 7-5

Pharmacodynamic Properties and Cardiac Effects of β-Adrenergic–Blocking Drugs









































































































































































































































DRUG RELATIVE β1 SELECTIVITY * ISA MSA RESTING HEART RATE EXERCISE HEART RATE RESTING MYOCARDIAL CONTRACTILITY RESTING BLOOD PRESSURE EXERCISE BLOOD PRESSURE RESTING AV CONDUCTION ANTIARRHYTHMIC EFFECT
Acebutolol + + + ↓– +
Atenolol ++ 0 0 +
Betaxolol ++ 0 + +
Bisoprolol ++ 0 0 +
Carteolol 0 + 0 ↓– ↓– +
Carvedilol 0 0 ++ ↓– ↓– ↓– +
Esmolol ++ 0 0 +
Labetalol § 0 + 0 ↓– ↓– ↓↓ ↓– +
Metoprolol ++ 0 0 +
Nadolol 0 0 0 +
Nebivolol ++ 0 0 ↓– +
Oxprenolol 0 + + ↓– ↓– ↓– +
Penbutolol 0 + 0 ↓– ↓– ↓– +
Pindolol 0 ++ + ↓– ↓– ↓– +
Propranolol 0 0 ++ +
Isomer-d-propranolol 0 0 ++ ↓– ↓– +
Sotalol 0 0 0 +
Timolol 0 0 0 +

AV, atrioventricular; ISA, intrinsic sympathomimetic activity; MSA, membrane stabilizing activity; ++, strong effect; +, modest effect; 0, absent effect; ↓, reduction; –, no change.

Modified from Frishman WH. Clinical pharmacology of the β-adrenoceptor blocking drugs, ed 2, Norwalk, CT, 1984, Appleton-Century-Crofts, p 15.

* β1 selectivity is seen only with low therapeutic drug concentrations. With higher concentrations, β1 selectivity is not seen.


Bisoprolol is also approved as a first-line antihypertensive therapy in combination with a very-low-dose diuretic.


Carvedilol has peripheral vasodilating activity and additional α1-adrenergic–blocking activity.


§ Labetalol has additional α1-adrenergic–blocking activity and direct vasodilatory activity.


Nebivolol has additional actions to increase endothelium-dependent vasodilation by increasing the activity of nitric oxide.


Effects of d-propranolol with doses in humans well above the therapeutic level; the isomer also lacks β-blocking activity.



Although exercise tolerance improves with β-blockade, the increments in heart rate and BP with exercise are blunted, and the rate/pressure product (systolic BP multiplied by heart rate) achieved when pain occurs is lower than that reached during a control run. The depressed rate/pressure product at the onset of pain, about 20% reduction from control, is reported to occur with various β-blockers, probably related to decreased cardiac output and possibly to a decrease in coronary perfusion. Thus, although exercise tolerance is increased with β-blockade, patients exercise less than might be expected. This may also relate to the action of β-blockers in increasing LV size, causing increased LV wall tension and an increase in oxygen consumption at a given BP.


Comparison with Other Antianginal Therapies


In a meta-analysis of clinical trial experience over 20 years that compared β-blockers, CCBs, and nitrates in patients who had stable angina pectoris, it was demonstrated that β-blockers provide an equivalent reduction in angina and lead to similar or reduced rates of adverse experiences compared with either CCBs or long-acting nitrates. The rates of cardiac death and MI were not significantly different for β-blockers than for CCBs.


Angina at Rest and Vasospastic Angina


Unstable angina pectoris can be caused by multiple mechanisms, including coronary vasospasm, myocardial bridging, and thrombosis, which appear to be responsible for ischemia in a significant proportion of patients with unstable angina and angina at rest. Therefore, because β-blockers primarily reduce myocardial oxygen consumption but fail to exert vasodilating effects on coronary vasculature, they may not be totally effective in patients whose angina is caused or increased by dynamic alterations in coronary luminal diameter. Despite potential dangers in rest and vasospastic angina, β-blockers have been used successfully as monotherapy and in combination with vasodilating antianginal agents in the majority of patients. In addition, there is evidence that β-blockers can reduce C-reactive protein levels, an inflammatory marker of increased CV morbidity and mortality.


Combined Use of β-Blockers with Other Antianginal Therapies in Angina Pectoris


Nitrates


As noted earlier, combined therapy with nitrates and β-blockers may be more efficacious for the treatment of angina pectoris than the use of either drug alone. The primary effects of β-blockers are to cause a reduction in both resting heart rate and the response of heart rate to exercise. Because nitrates produce a reflex increase in heart rate and contractility from a reduction in arterial pressure, concomitant β-blocker therapy is extremely effective because it blocks this reflex increment in the heart rate. Similarly, the preservation of diastolic coronary blood flow with a reduced heart rate will also be beneficial. In patients with a propensity for myocardial failure who may have a slight increase in heart size with the β-blockers, the nitrates will counteract this tendency by reducing heart size as a result of its peripheral venodilator effects. During the administration of nitrates, the reflex increase in contractility mediated through the sympathetic nervous system will be blunted by the presence of β-blockers. Similarly, the increase in coronary resistance associated with β-blocker administration can be ameliorated by the administration of nitrates.


Calcium Channel Blockers


Some CCBs (diltiazem, verapamil) also slow the heart rate and inhibit AV nodal conduction. Combined therapy with β-blockers and CCBs can provide clinical benefits for patients with angina pectoris who remain symptomatic with the use of either agent alone. Because adverse CV effects can also occur with combination treatment, such as heart block and excessive myocardial depression, patients being considered for such treatment must be carefully selected and observed.


Hemodynamically, these two types of agents have different effects on the circulation (see Tables 7-3 and 7-5 ), leading to the possibility of therapeutic combination. Of the combinations, β-blockade plus a DHP such as nifedipine is likely to be simplest. The DHPs do not inhibit the SA or AV node and therefore can be more readily combined with a β-blocker than can the NDHPs, such as verapamil and diltiazem. Because the tendency to produce tachycardia with the DHPs is antagonized by the β-blocker, there are no additive effects on the SA or AV node. Through vasodilation, including coronary vasodilation, the DHPs can contribute to the antianginal effect. β-Blockade should be combined with the NDHPs, such as verapamil and diltiazem, only after consideration of the risks and after plans are in place for patient monitoring. With NDHP CCBs comes the risk of extreme bradycardia, AV nodal block, or a marked negative inotropic effect. Second-generation CCBs—such as the DHPs amlodipine, felodipine, isradipine, and nicardipine—can also be readily combined with β-blockade.


Ranolazine


Ranolazine is a piperazine derivative that is approved in an extended-release tablet as a first-line treatment for chronic angina pectoris. The drug can also be combined with β-blockers to provide additional antianginal relief, but it is not approved for use in unstable angina.


Conditions Associated with Angina Pectoris


Arrhythmias


β-Blockers are an important treatment modality for various cardiac arrhythmias, especially in patients with ischemic heart disease. Although it was initially believed that β-blockers were more effective in treating supraventricular arrhythmias than ventricular arrhythmias, subsequent studies suggest that this may not be the case. β-Blockers can be quite useful in the prevention and treatment of ventricular tachyarrhythmias in the setting of myocardial ischemia, mitral valve prolapse, the hereditary QT-interval prolongation syndrome, and other CV conditions, such as cardiomyopathy. β-Blockers can be combined with amiodarone with relative safety and synergy of antiarrhythmic action and with implantable cardioverter-defibrillators to reduce the frequency of shocks.


Hypertension


The mechanism of the antihypertensive effect of β-blockade is still under dispute, but its effect on overall CV mortality appears to be similar to other classes of antihypertensive drugs. Initially, β-blockers decrease the heart rate and cardiac output falls by about 20%, yet the BP does not fall because the arteriolar resistance reflexively increases. Within 24 hours of the start of β-blocker treatment, the peripheral resistance starts to fall, so arterial pressure declines. The mechanism of this delayed hypotensive effect is unclear, but it is thought to involve inhibition of prejunctional β-adrenergic receptors. Alternatively, inhibition of the renin-angiotensin system may account for the delayed vasodilation. Additional antihypertensive mechanisms may involve a central action and decreased renin release.


Survivors of Acute Myocardial Infarction


β-Blockers have beneficial effects on many determinants of myocardial ischemia (see Box 7-1 and Chapters 9 and 10 ). The results of placebo-controlled, long-term treatment trials with some β-blockers in survivors of acute MI demonstrated a favorable effect on total mortality rates; CV mortality rates, including sudden and nonsudden cardiac deaths; and the incidence of nonfatal reinfarction. Patients in these studies included those who had relative contraindications to β-blockade but still appeared to benefit and in patients with diabetes, who also responded favorably to treatment. The beneficial results of β-blocker therapy can be explained by both the antiarrhythmic and antiischemic effects of these drugs. It has also been proposed that β-blockers reduce the risk of atherosclerotic plaque fissure and subsequent thrombosis. Two nonselective β-blockers, propranolol and timolol, are approved for use in reducing the risk of death in MI survivors when started 5 to 28 days after an MI. Metoprolol and atenolol, two β1-selective blockers, are approved for the same indication, and both can be used intravenously in the hyperacute phase of an MI. β-Blockers have also been suggested as a treatment to reduce the extent of myocardial injury and deaths during the hyperacute phase of MI. The α-/β-blocker carvedilol is indicated to reduce CV mortality in clinically stable patients who have survived the acute phase of MI and have an LV ejection fraction of less than 40% with or without symptomatic heart failure. IV and oral atenolol have been shown to be effective in causing a modest reduction in early mortality rates when administered during the hyperacute phase of acute MI. Atenolol and metoprolol reduce early infarct mortality rates by 15%, an effect that may be improved when β-blockade is combined with thrombolytic therapy. Despite all of the evidence showing that β-blockers are beneficial in patients who survive MI, they are still considerably underused in clinical practice. β-Blockers should not be administered in patients who come to medical attention with MI and have evidence of heart failure, low cardiac output, or increased risk of cardiogenic shock.


“Silent” Myocardial Ischemia


Investigators have observed that not all myocardial ischemic episodes detected on ECG are associated with detectable symptoms. Positron emission imaging techniques have validated the theory that these silent ischemic episodes are indicative of true myocardial ischemia. Compared with symptomatic ischemia, the prognostic importance of silent myocardial ischemia that occurs at rest or during exercise has not been determined.


β-Blockers are as successful in reducing the frequency and timing of silent ischemic episodes detected by ambulatory ECG monitoring as they are in reducing the frequency of painful ischemic events.


Other Cardiovascular Conditions Associated with Angina Pectoris


Although β-blockers have been studied extensively in patients with angina pectoris, arrhythmias, and hypertension, they have also been shown to be safe for other CV conditions associated with angina pectoris.


Hypertrophic Cardiomyopathy


β-Blockers without partial agonist activity have been proven effective for patients with hypertrophic cardiomyopathy. These drugs are useful for reducing dyspnea, angina, and syncope. β-Blockers have also been shown to lower the intraventricular pressure gradient both at rest and with exercise.


The outflow pressure gradient is not the only abnormality in hypertrophic cardiomyopathy; more important is the loss of ventricular compliance, which impedes normal LV function. It has been shown through both invasive and noninvasive methods that propranolol can improve LV function in this condition. The drug also produces favorable changes in ventricular compliance while it relieves symptoms. Propranolol has been approved for this condition and may be combined with the CCB verapamil or disopyramide in patients who do not respond to the β-blocker alone.


The salutary hemodynamic and symptomatic effects produced by β-blockers derive from their inhibition of sympathetic stimulation of the heart. No evidence suggests that the drug alters the primary cardiomyopathic process; many patients remain in or return to their severely symptomatic state, and some patients die despite β-blocker administration.


Congestive Cardiomyopathy


The ability of IV sympathomimetic amines to effect an acute increase in myocardial contractility through stimulation of the β-adrenergic receptor had prompted the hope that the use of oral catecholamine analogs could provide long-term benefit for patients with severe heart failure. However, observations concerning the regulation of the myocardial adrenergic receptor and abnormalities of β-adrenergic receptor–mediated stimulation of the failing myocardium have caused a critical reappraisal of the scientific validity of sustained β-adrenergic receptor stimulation. Evidence suggests that β-adrenergic receptor blockade, when tolerated, may have a favorable effect on the underlying cardiomyopathic process.


Enhanced sympathetic activation is seen consistently in patients with CHF and is associated with decreased exercise tolerance, hemodynamic abnormalities, and increased mortality rates. Increases in sympathetic tone can potentiate the renin-angiotensin system in patients and lead to increased salt and water retention, arterial and venous constriction, and increments in ventricular preload and afterload. Elevated levels of catecholamines can increase heart rate and cause coronary vasoconstriction, adversely influence myocardial contractility on the cellular level, and cause myocyte hypertrophy and vascular remodeling. Catecholamines can stimulate growth and provoke oxidative stress in terminally differentiated cardiac cells; these two factors can trigger the process of programmed cell death known as apoptosis. Finally, excess catecholamines can increase the risk of sudden death in patients with CHF by adversely influencing the electrophysiologic properties of the failing heart.


Controlled trials with several β-blockers in patients with either ischemic or nonischemic cardiomyopathy showed that these drugs improve symptoms, ventricular function, and functional capacity while reducing the need for hospitalization. A series of placebo-controlled clinical trials with the α-/β-blocker carvedilol and the β1-selective agents bisoprolol and metoprolol have shown a mortality benefit in patients with New York Heart Association (NYHA) functional class II to IV heart failure, when the drug was used in addition to diuretics, ACE inhibitors, and digoxin. For patients with class II to III heart failure, initial treatment with a β-blocker followed by an ACE inhibitor was found to be at least as effective as beginning with an ACE inhibitor.


The mechanisms of benefit with β-blocker use are not yet known. Possible mechanisms for β-blocker benefit in chronic heart failure include the upregulation of impaired β-adrenergic receptor expression in the heart and an improvement in impaired baroreceptor functioning, an effect that can inhibit excess sympathetic outflow. It has been suggested that long-term therapy with β-blockers improves the left atrial contribution to LV filling while increasing the levels of cardiac natriuretic peptides.


Mitral Valve Prolapse


Atypical chest pain, malignant arrhythmias, and nonspecific ST- and T-wave abnormalities have been observed with this condition. By decreasing sympathetic tone, β-blockers have been shown to be useful for relieving the chest pains and palpitations that many of these patients experience and for reducing the incidence of life-threatening arrhythmias and other ECG abnormalities.


Dissecting Aneurysms


β-Blockade plays a major role in the treatment of patients with acute aortic dissection. During the hyperacute phase, β-blockers reduce the force and velocity of myocardial contraction (dP/dT) and hence slow the progression of the dissecting hematoma. Moreover, β-blockade should be initiated simultaneously with the institution of other antihypertensive therapy (e.g., sodium nitroprusside) that may cause reflex tachycardia and increases in cardiac output, factors that can aggravate the dissection process. β-Blockade is administered intravenously to reduce the heart rate to less than 60 beats/min. Once a patient is stabilized—that is, once adequate control of heart rate and BP has been achieved and no further pain from dissection is apparent—and long-term medical management is contemplated, the patient should be maintained on oral β-blocker therapy to prevent the recurrence of dissection.


Ehlers-Danlos Syndrome


In a placebo-controlled study, the long-term use of a β-blocker has been shown to reduce the risk of spontaneous rupture of the aorta in the vascular subtype of Ehlers-Danlos syndrome.


Syndrome X


A dysfunction of small coronary arterial vessels has been hypothesized to be responsible for syndrome X, a chest pain syndrome that often occurs without evidence of large-vessel CAD. The treatment of syndrome X, however, remains largely empiric and is often unsatisfactory. Some investigators found that β-blockers, rather than CCBs and nitrates, were useful in relieving symptoms, suggesting that they may be the preferred drugs when starting pharmacologic treatment for syndrome X.


Perioperative Therapy in High-Risk Patients with Ischemic Heart Disease


β-Adrenergic drugs will reduce the risk of perioperative ischemia and arrhythmias. Based on these studies, several national organizations have endorsed the perioperative use of β-blockers as a best practice. However, some recent evidence would suggest that the routine use of β-blockers may actually cause harm in some patients. Currently, the best available evidence supports their use in two patient groups: 1) those undergoing vascular surgery who have known ischemic heart disease or multiple risk factors for it and 2) those undergoing vascular surgery who are already receiving β-blockers for cardiovascular conditions. When feasible, β-blockers should be started 1 month before cardiac surgery, with the dose titrated to achieve a heart rate of 60 beats/min, and should be continued for 1 month after surgery.


Pharmacologic Differences Among β-Adrenergic Receptor–Blocking Drugs


More than 100 β-blockers have been synthesized, and more than 30 are available worldwide for clinical use. Selectivity for two subgroups of the β-adrenergic–receptor population has been prominent in the development of β-blockers: β1-adrenergic receptors in the heart and β2-adrenergic receptors in the peripheral circulation and bronchi. More controversial has been the introduction of β-blockers with α-adrenergic receptor–blocking actions, varying amounts of selective and nonselective intrinsic sympathomimetic activity (partial agonist activity), CCB activity, nitric oxide potentiating action, and nonspecific membrane-stabilizing effects ( Table 7-6 ). Pharmacokinetic differences also exist among β-blockers that may be of clinical importance.



TABLE 7-6

Properties of Various β-Adrenoceptor Antagonist Agents: Noncardioselective vs. Cardioselective and Vasodilatory Agents













































































































































































































Drug ISA PLASMA HALF-LIFE (houRS) LIPID SOLUBILITY * FIRST-PASS EFFECT LOSS BY LIVER/KIDNEY PLASMA PROTEIN BINDING (%) USUAL DOSE FOR ANGINA (OTHER INDICATIONS) USUAL DOSE AS SOLE THERAPY FOR MILD/MODERATE HYPERTENSION
Noncardioselective
Propranolol (Inderal, Innopran) 1-6 +++ ++ Liver 90 80 mg bid usually adequate (may give 160 mg bid) Start with 10 to 40 mg bid, mean 160 to 320 mg/day, 1 or 2 doses
Inderal LA 8-11 +++ ++ Liver 90 80-320 mg qd 80-320 mg daily
Innopran XL 8-11 +++ ++ Liver 90 Not indicated 80-120 mg at bedtime
Carteolol (Cartrol) + 5-6 0/+ 0 Kidney 20-30 Not evaluated 2.5-10 mg single dose
Nadolol (Corgard) 20-24 0 0 Kidney 30 40-80 mg qd up to 240 mg 40-80 mg/day up to 320 mg
Penbutolol (Levatol) + 20-25 +++ ++ Liver 98 Not studied 10-20 mg/day
Sotalol (Betapace, Betapace AF) § 7-18, mean 12 0 0 Kidney 5 80-240 mg bid in 2 doses for serious ventricular arrhythmias; up to 160 mg bid for atrial fibrillation, flutter 80-320 mg/day, mean 190 mg/day
Timolol (Blocadren) 4-5 + + Liver, kidney 60 10 mg bid after MI 10-20 mg bid
Cardioselective
Acebutolol (Sectral) ++ 8-13 0 Liver, kidney 15 400-1200 mg/day in 2 doses for PVCs 400-200 mg/day; can be given as a single dose
Atenolol (Tenormin) 6-7 0 0 Kidney 10 50-200 mg qd 50-100 mg/day
Betaxolol (Kerlone) 14-22 ++ ++ Liver, kidney 50 10-20 mg/day
Bisoprolol (Zebeta) 9-12 + 0 Liver, kidney 30 10 mg qd (not in the United States) 2.5-40 mg/day
Metoprolol (Lopressor, Toprol) 3-7 + ++ Liver 12 50-200 mg bid 100-400 mg/day in 1 or 2 doses
Toprol-XL Slow release + ++ Liver 12 100-400 mg qd As above, 1 dose
Vasodilatory β-Blockers
Noncardioselective
Labetalol (Trandate, Normodyne) 6-8 +++ ++ Liver, some kidney 90 As for hypertension 300-600 mg/day in 3 doses; top dose 2400 mg/day
Pindolol (Visken) β1, β2 4 + + Liver, kidney 55 2.5-7.5 mg tid (not in the United States) 5-30 mg in 2 daily doses
Carvedilol (Coreg) 6 + ++ Liver 95 In the United States and United Kingdom, licensed for heart failure up to 25 mg bid; start with low dose 12.5-25 mg bid
Cardioselective
Nebivolol (Bystolic) 6-10 ++ ++ Liver, kidney 98 2.5-10 mg qd 2.5-10 mg qd

ISA, intrinsic sympathomimetic activity; MI, myocardial infarction; PVC, premature ventricular contraction; +++, very strong effect; ++, strong effect; +, modest effect; 0, absent effect; –, negative effect.

Modified from Opie LH, Yusuf S. Beta-blocking agents. In Opie LH, Gersh BJ, editors: Drugs for the heart, 5th ed. Philadelphia, 2001, WB Saunders, pp 1-32.

* Octanol-water distribution coefficient (pH 7.4, 37° C), where 0 is ≤0.5, + is 0.5 to 2, ++ is 2 to 10, and +++ is ≥10.


Approved by the FDA for hypertension.


Approved by the FDA for angina pectoris.


§ Approved by the FDA for life-threatening ventricular tachyarrhythmias.


Approved by the FDA for heart failure.



Sixteen β-blockers are marketed in the United States for CV disorders: propranolol for angina pectoris, arrhythmias, systemic hypertension, migraine prophylaxis, essential tremor, hypertrophic cardiomyopathy, and reduction in the risk of CV death in survivors of an acute MI; nadolol for hypertension and angina pectoris; timolol for hypertension and to reduce the risk of CV death and nonfatal reinfarction in survivors of MI and for a topical form for glaucoma; atenolol and metoprolol for hypertension and angina and in IV and oral formulations to reduce the risk of CV death in survivors of MI; penbutolol, bisoprolol, nebivolol, pindolol, and carvedilol for hypertension; betaxolol and carteolol for hypertension and in a topical form for glaucoma; acebutolol for hypertension and ventricular arrhythmias; IV esmolol for supraventricular arrhythmias; sotalol for atrial and ventricular arrhythmias; and labetalol for hypertension and in an IV form for hypertensive emergencies. Carvedilol, metoprolol, and bisoprolol are approved for clinical use in the treatment of CHF.


Despite extensive experience with β-blockers in clinical practice, no studies have been done that suggest any of these agents provides major advantages or disadvantages compared with the others for the treatment of many CV diseases. When any available β-blocker is titrated properly, it can be effective in patients with arrhythmia, hypertension, or angina pectoris (see Table 7-6 ). However, one agent may be more effective than other agents in reducing adverse reactions in some patients and in managing specific situations.


Potency


β-Blockers are competitive inhibitors of catecholamine binding at β-adrenergic receptor sites. The dose-response curve of the catecholamine is shifted to the right; that is, a given tissue response requires a higher concentration of agonist in the presence of β-blockers. β1-Blocking potency can be assessed by the inhibition of tachycardia produced by isoproterenol or exercise (the more reliable method in the intact organism), and potency varies among compounds. These differences in potency are of no therapeutic relevance, but they do explain the different drug doses needed to achieve effective β-blockade when initiating therapy in patients or when switching from one agent to another.


β1 Selectivity


β-Blockers may be classified as selective or nonselective based on their relative ability to antagonize the actions of sympathomimetic amines in some tissues at lower doses than those required in other tissues. When used in low doses, β1-selective blockers such as acebutolol, betaxolol, bisoprolol, esmolol, atenolol, and metoprolol inhibit cardiac β1-adrenergic receptors but have less influence on bronchial and vascular β-adrenergic receptors (β2). In higher doses, however, β1-selective blockers also block β2-adrenergic receptors. Accordingly, β1-selective agents may be safer than nonselective agents in patients with obstructive pulmonary disease, because β2-adrenergic receptors remain available to mediate adrenergic bronchodilation. Even relatively selective β-blockers may aggravate bronchospasm in certain patients, so these drugs should generally not be used in patients with active bronchospastic disease.


A second theoretical advantage is that, unlike nonselective β-blockers, β1-selective blockers in low doses may not block the β2-adrenergic receptors that mediate the dilation of arterioles. During the infusion of epinephrine, nonselective β-blockers can cause a pressor response by blocking β2-adrenergic receptor–mediated vasodilation, because β-adrenergic vasoconstrictor receptors are still operative. Selective β1-blockers may not induce this pressor effect in the presence of epinephrine and may lessen the impairment of peripheral blood flow. It is possible that leaving the β2-adrenergic receptors unblocked and responsive to epinephrine may be functionally important in some patients with asthma, hypoglycemia, hypertension, or peripheral vascular disease when they are treated with β-blockers.


Intrinsic Sympathomimetic Activity (Partial Agonist Activity)


Certain β-blockers have an intrinsic sympathomimetic activity (partial agonist activity) at β1-adrenergic receptor sites, β2-adrenergic receptor sites, or both. In a β-blocker, this property is identified as a slight cardiac stimulation that can be blocked by propranolol. The β-blockers with this property partially activate the β-adrenergic receptor in addition to preventing the access of natural or synthetic catecholamines to the receptor. Dichloroisoprenaline, the first β-adrenergic receptor-blocking drug to be synthesized, exerted such marked partial agonist activity that it was unsuitable for clinical use. However, compounds with less partial agonist activity are effective β-blockers. The partial agonist effects of β-blockers such as pindolol differ from those of agonists epinephrine and isoproterenol in that the maximum pharmacologic response that can be obtained is low, although the affinity for the receptor is high. In the treatment of patients with arrhythmias, angina pectoris of effort, and hypertension, drugs with mild to moderate partial agonist activity appear to be as efficacious as β-blockers that lack this property. It is still debated whether the presence of partial agonist activity in a β-blocker constitutes an overall advantage or disadvantage in cardiac therapy. Drugs with partial agonist activity cause less slowing of the heart rate at rest than do propranolol and metoprolol, although the increments in heart rate with exercise are similarly blunted. These β-blockers reduce peripheral vascular resistance and may cause less depression or AV conduction than drugs that lack these properties. Some investigators claim that partial agonist activity in a β-blocker protects against myocardial depression, adverse lipid changes, bronchial asthma, and peripheral vascular complications, such as those caused by propranolol.


The evidence to support these claims is not conclusive, and more definitive clinical trials will be necessary to resolve these issues.


α-Adrenergic Activity


Labetalol is a β-blocker with antagonistic properties at both α- and β-adrenergic receptors, and it has direct vasodilator activity. Labetalol has been shown to be 6 to 10 times less potent than phentolamine at α-adrenergic receptors, 1.5 to 4 times less potent than propranolol at β-adrenergic receptors, and is itself 4 to 16 times less potent at α- than at β-adrenergic receptors. Like other β-blockers, it is useful in the treatment of hypertension and angina pectoris. Unlike most β-blockers, however, the additional α-adrenergic receptor–blocking actions of labetalol lead to a reduction in peripheral vascular resistance that may maintain cardiac output. Whether concomitant α-adrenergic receptor–blocking activity is actually advantageous in a β-blocker remains to be determined.


Carvedilol is another β-blocker with additional β-adrenergic receptor–blocking activity, with an α1- to β-blockade ratio of 1 : 10. On a milligram/milligram basis, carvedilol is about 2 to 4 times more potent than propranolol as a β-blocker. In addition, carvedilol may have antioxidant and antiproliferative activities, it has been used for the treatment of hypertension and angina pectoris, and it is approved as a treatment for hypertension and for patients with symptomatic heart failure.


Nitric Oxide Potentiating Effect


A novel aspect of the pharmacology of the β1-selective antagonist nebivolol is its ability to produce endothelium-dependent vasodilation through a nitric oxide pathway. Nebivolol produces vasodilation by acting as a β3-receptor agonist, which increases the activity of nitric oxide. Nitric oxide activity is also augmented by nebivolol through the prevention of nitric oxide deactivation. The nitric oxide–mediated vasodilatory effects of nebivolol occur primarily in the small arteries and contribute to the effect of the drug on arterial BP.


Pharmacokinetics


Although the β-blockers as a group have similar therapeutic effects, their pharmacokinetic properties are markedly different. Their varied aromatic ring structures lead to differences in completeness of GI absorption, amount of first-pass hepatic metabolism, lipid solubility, protein binding, extent of distribution in the body, penetration into the brain, concentration in the heart, rate of hepatic biotransformation, pharmacologic activity of metabolites, and renal clearance of a drug and its metabolites, which may influence the clinical usefulness of these drugs in some patients. The desirable pharmacokinetic characteristics of β-blockers in general are a lack of major individual differences in bioavailability and in metabolic clearance of the drug and a rate of removal from active tissue sites that is slow enough to allow longer dosing intervals.


The β-blockers can be divided by their pharmacokinetic properties into two broad categories: those eliminated via hepatic metabolism, which tend to have relatively short plasma half-lives, and those eliminated unchanged by the kidney, which tend to have longer half-lives. Propranolol and metoprolol are both lipid soluble, are almost completely absorbed by the small intestine, and are largely metabolized by the liver. They tend to have more variable bioavailability and relatively short plasma half-lives. A lack of correlation between the duration of clinical pharmacologic effect and plasma half-life may allow these drugs to be administered once or twice daily.


In contrast, agents such as atenolol and nadolol are more water soluble, are incompletely absorbed through the gut, and are eliminated unchanged by the kidney. They tend to have less variable bioavailability in patients with normal renal function in addition to longer half-lives, which allows once-a-day dosing. The longer half-lives may be useful in patients who find compliance with frequent β-blocker dosing problematic.


Long-acting sustained-release preparations of propranolol and metoprolol are available. Studies have shown that long-acting propranolol and metoprolol can provide a much smoother curve of daily plasma levels than can be comparable to divided doses of conventional immediate-release formulations. In addition, a delayed-release/sustained-release formulation of propranolol is available that is designed to target early morning elevations in BP and heart rate related to circadian rhythms.


The specific pharmacokinetic properties of individual β-blockers—first-pass metabolism, active metabolites, lipid solubility, and protein binding—may be clinically important. When drugs with extensive first-pass metabolism are taken by mouth, they undergo so much hepatic biotransformation that relatively little drug reaches the systemic circulation. Depending on the extent of first-pass effect, an oral dose of β-blocker must be larger than an IV dose to produce the same clinical effects. Some β-blockers are transformed into pharmacologically active compounds (e.g., acebutolol) rather than inactive metabolites. The total pharmacologic effect depends on the amount of the drug administered and its active metabolites. Characteristics of lipid solubility in a β-blocker have been associated with the ability of the drug to concentrate in the brain, and many side effects of these drugs—such as lethargy, mental depression, and hallucinations—that have not been clearly related to β-blockers may result from their actions on the central nervous system. However, it is still uncertain whether drugs that are less lipid soluble cause fewer of these adverse reactions.


Some genetic polymorphisms can influence the metabolism of various β-blockers, including propranolol, metoprolol, timolol, and carvedilol. A one-codon difference of CYP 2D6 may explain a significant proportion of interindividual variation in the pharmacokinetics of propranolol in Chinese subjects. In addition, exercise has not been shown to have any effect on the pharmacokinetics of propranolol.


Adverse Effects of β-Adrenergic Receptor Blockers


An evaluation of adverse effects is complex because of the use of different definitions of side effects, the kinds of patients studied, study design features, and different methods of ascertaining and reporting adverse side effects among studies. Overall, the types and frequencies of adverse effects attributed to various β-blocker compounds appear similar. The side-effect profiles resemble those seen with concurrent placebo treatments, attesting to the remarkable safety margin of β-blockers.


Adverse effects of β-blockers are an exaggeration of the normal cardiac therapeutic effects, resulting in excess bradycardia, AV nodal block, and excess negative inotropic effect. All β-blockers tend to promote bronchospasm, with low doses of β1-selective agents being the least harmful. Cold extremities occur with both selective and nonselective agents, yet agents with intrinsic sympathomimetic activity may provide a slightly better skin temperature than propranolol, at least during an acute study. The adverse effects of all β-blockers on the peripheral circulation may be less marked than previously thought.


Fatigue is a frequent side effect, again found particularly with propranolol, with less of an effect when a β1-selective or vasodilatory blocker is used, so both central and peripheral hemodynamic mechanisms may be involved. Although one double-blind study shows no difference between the effects of the β1-selective agent atenolol and placebo, exercise physiologists find that some impairment in peak exercise occurs with all β-blockers.


Impotence is often reported by patients who receive β-blockers, usually middle-aged men with atherosclerotic arterial disease. In one study, erectile dysfunction occurred in 11% of patients administered a β-blocker for hypertension compared with 26% of these patients administered a diuretic and 3% of placebo-treated patients.


An impaired quality of life found especially with propranolol is theoretically ascribed to its lipid solubility and brain penetration. Yet a variety of β-blockers other than propranolol, and with different pharmacologic properties, preserve quality of life in hypertensive patients. Central effects of β-blockers are often subtle and are not always explicable by the lipid-penetration hypothesis.


β-Blockers have effects on various metabolic parameters, including blood glucose and blood lipids. In a prospective cohort study of 12,550 nondiabetic individuals with hypertension, β-blockers were shown to increase the risk of developing type 2 diabetes, a finding not observed with thiazide diuretics, ACE inhibitors, or CCBs. This increased risk of diabetes must be weighed against the proven benefits of β-blockers in reducing the risk of CV events in patients with ischemic heart disease. Studies are needed to determine whether the use of ACE inhibitors in conjunction with β-blockers might counteract the adverse effects of β-blockers with respect to glucose tolerance. Carvedilol has been shown not to affect glycemic control, and it improves some components of the metabolic syndrome relative to metoprolol in diabetic patients.


Similarly, β-blockers without intrinsic sympathomimetic activity have been shown in hypertensive patients to decrease high-density lipoprotein cholesterol concentrations by 7% to 10% and to raise triglyceride concentrations by 10% to 20%. These small changes in lipids induced by β-blockers do not appear to diminish the beneficial effects of BP lowering on morbidity and mortality rates from coronary heart disease and stroke.


Contraindications to β-Adrenergic Receptor Blockers


Several absolute contraindications exist, which include CV contraindications such as severe bradycardia (heart rate <40 beats/min); preexisting high-degree AV nodal block (PR interval of >0.24 seconds without a functioning pacemaker); overt LV failure, except when the β-blocker is administered initially at low doses and under supervision to patients already receiving diuretics, digoxin, and an ACE inhibitor; and active peripheral vascular disease with rest ischemia. Severe bronchospasm is an absolute contraindication, even to β-selective agents; severe psychological depression is an important relative contraindication, particularly for propranolol.


Overdosage


Suicide attempts and accidental overdosing with β-blockers are being described with increasing frequency. Because β-blockers are competitive pharmacologic antagonists, their life-threatening effects—bradycardia, myocardial failure, and ventilatory failure—can be overcome with an immediate infusion of a β-agonist agent such as isoproterenol or dobutamine. When catecholamines are not effective, IV glucagon, amrinone, or milrinone has been used. There are no published recommended doses of IV catecholamines or phosphodiesterase inhibitors to treat β-blocker overdose; such agents should be used in their usual pharmacologic concentration until it is certain that reversal of β-blocker toxicity—reversal of heart blocks, excessive bradycardia, and myocardial depression—has occurred.


Monitoring of cardiorespiratory function is necessary for at least 24 hours in an intensive care unit after the patient responds to treatment of the β-blocker overdose. Patients who recover usually have no long-term sequelae; however, they should be observed for the cardiac signs of sudden β-blocker withdrawal.


β-Adrenergic Receptor Blocker Withdrawal


After abrupt cessation of long-term β-blocker therapy, exacerbation of angina pectoris and, in some cases, acute MI and death have been reported. Observations made in multiple double-blind randomized trials have confirmed the reality of a propranolol withdrawal reaction. The mechanism for this reaction is unclear, but some evidence suggests that the withdrawal phenomenon may be due to the generation of additional β-adrenergic receptors during the period of β-blockade. When the β-blocker is then withdrawn, the increased β-adrenergic receptor population readily results in excessive β-adrenergic receptor stimulation, which is clinically important when the delivery and use of oxygen are finely balanced, as occurs in ischemic heart disease. Other suggested mechanisms for the withdrawal reaction include heightened platelet aggregability, an elevation in thyroid hormone activity, and an increase in circulating catecholamines. Similar withdrawal problems have been seen with β-blocker discontinuation in patients with heart failure previously responsive to treatment.


Drug-Drug Interactions


β-Blockers are commonly used with other cardiovascular and noncardiovascular drugs, and the list of drugs with which they interact is extensive ( Table 7-7 ). The majority of the reported interactions have been associated with propranolol, the best-studied β-blocker, and such findings may not necessarily apply to other drugs in this class.


Mar 21, 2019 | Posted by in GENERAL | Comments Off on Pharmacologic Options for Treatment of Ischemic Disease

Full access? Get Clinical Tree

Get Clinical Tree app for offline access