Alpha-blockers should not be used as first-line therapy for hypertension. However, an alpha-blocker can be considered as a second-line or third-line add-on in a patient whose blood pressure is not under control despite treatment with other drugs.
In addition, alpha-blockers are useful in relieving lower urinary tract symptoms in patients with benign prostatic hypertrophy. However, even in a patient who has both hypertension and benign prostatic hypertrophy, we advise physicians to use alpha-blockers primarily to relieve the urinary symptoms, and we recommend lowering the blood pressure with a drug of a class shown to reduce rates of illness and death.
NOT FIRST-LINE THERAPY
All antihypertensive drugs, including alpha-blockers, lower blood pressure. Alpha-blockers have been approved by the US Food and Drug Administration for treating high blood pressure, and they are just as effective as other antihypertensive drugs—if efficacy is defined as a decrease in millimeters of mercury.
However, lowering the blood pressure is not the main goal of antihypertensive therapy. What we want to achieve when prescribing antihypertensive drugs is to reduce the rates of heart attacks, strokes, and other adverse cardiovascular adverse outcomes, including death.
Unfortunately, alpha-blockers fall short in this regard. In the Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack (ALLHAT) trial,1,2 doxazosin (Cardura) was found to carry a higher risk of combined cardiovascular disease (relative risk 1.19, P = .04), mostly stroke. Alarmingly, the incidence of symptomatic heart failure in patients on doxazosin was twice that in patients on chlorthalidone (relative risk 2.04, P < .001). Doxazosin was minimally more effective in lowering blood pressure than chlorthalidone, but the small difference in blood pressure was unlikely to have accounted for the significant difference in the risk of heart failure.3
This experience with doxazosin illustrates a key drawback to surrogate end points: a treatment may produce a favorable outcome in the surrogate end point (blood pressure) but produce little or no benefit in terms of the real end point (stroke, myocardial infarction, and heart failure).4
Based on the ALLHAT data as well as on a Veterans Administration study in patients with chronic heart failure in which survival with prazosin (Minipress) was no better than with placebo,5 it seems reasonable to no longer use alpha-blockers as initial therapy for hypertension. This view is reflected by current European6 and American7 guidelines.
ALPHA-BLOCKERS AS PART OF COMBINATION THERAPY
In several clinical trials, alpha-blockers were allowed8 or were specified9,10 as add-on therapy if other drugs failed to control the blood pressure, but they were not used in a randomized fashion. Thus, we cannot judge their effect on cardiovascular outcomes such as heart attack and stroke.
The choice of drugs for combination therapy very often is still empirical and based on personal preference. Doxazosin as add-on therapy, in general, has been shown to be safe and well tolerated.11 But even if it is acceptable, it is not a preferred combination.
In the Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT),9 patients received extended-release doxazosin as a third drug if they did not reach their goal blood pressure with either the combination of amlodipine (Norvasc) plus perindopril (Aceon) or atenolol (Tenormin) plus bendroflumethiazide. Extended-release doxazosin was an effective add-on, and there was no apparent excess rate of heart failure in doxazosin users.
In other studies, in patients with uncontrolled hypertension, adding doxazosin as a second- or third-line agent to a gold-standard drug—calcium channel blocker, diuretic, beta-blocker, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, or combinations of these—allowed significantly more participants to achieve their blood pressure goal.11
Personally, we consider doxazosin in patients whose blood pressure is not controlled with triple therapy with a renin-angiotensin system blocker, a diuretic, and a calcium channel antagonist in full doses. In patients with stage 3 or stage 4 kidney disease who can no longer tolerate renin-angiotensin system blockers, doxazosin may also be a useful adjunct. Whether the metabolic effects of alpha-blockers, such as a reduction in insulin resistance and a decrease in total and low-density lipoprotein cholesterol, will result in lower rates of morbidity and death has not been conclusively determined.
A point of view somewhat more favorable to the use of alpha-blockers has recently been put forward by Chapman et al.12
ALPHA-BLOCKERS ALLEVIATE SYMPTOMS OF BENIGN PROSTATIC HYPERTROPHY
Doxazosin and other alpha-blockers are commonly used to alleviate lower urinary tract symptoms in patients with benign prostatic hypertrophy.
Both high blood pressure and benign prostatic hypertrophy become more common with advancing age, and it has been estimated that both are present in more than 25% of men over age 60.13 Indeed, two trials documented that a significant reduction in symptoms of benign prostatic hypertrophy and in systolic and diastolic blood pressure can be achieved with an alpha-blocker.13,14
This raises the question whether such a “twofer” (treating two disease states with one drug) should be used in clinical practice. We have to consider that the principle of the twofer has never been tested and agree with Davis et al,3 who, in a further analysis of the ALLHAT data, stated that, “In older men with benign prostatic hypertrophy in whom an [alpha]-adrenergic blocker seems like the best treatment for the uropathy, coexisting hypertension should be treated with another antihypertensive drug as well.”3
Again, this would clearly relegate doxazosin to second-line or third-line status, even in patients with benign prostatic hypertrophy, in whom it has been shown to be indicated.
ADVERSE EFFECTS OF ALPHA-BLOCKERS
Dizziness, fatigue, and somnolence are occasionally reported but appear to be well tolerated. Postural hypotension is much less common with proper titration of standard doxazosin or with the use of controlled-release formulations.9–15 However, in patients with impaired autonomic function, even long-acting alpha-blockers can cause postural hypotension and syncope.
Patients using phosphodiesterase type 5 inhibitors—sildenafil (Viagra), vardenafil (Levitra), or tadalafil (Cialis)—for erectile dysfunction should avoid alpha-blockers because the blood-pressure-lowering effects of the two drug classes may be additive.
References
Messerli FH. Implications of discontinuation of doxazosin arm of ALLHAT. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (commentary). Lancet2000; 355:863–864.
Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). ALLHAT Collaborative Research Group. JAMA2000; 283:1967–1975.
Davis BR, Cutler JA, Furberg CD, et al; ALLHAT Collaborative Research Group. Relationship of antihypertensive treatment regimens and change in blood pressure to risk for heart failure in hypertensive patients randomly assigned to doxazosin or chlorthalidone: further analyses from the Antihypertensive and Lipid-Lowering treatment to prevent Heart Attack Trial. Ann Intern Med2002; 137:313–320.
Messerli FH. Doxazosin and congestive heart failure (viewpoint). J Am Coll Cardiol2001; 38:1295–1296.
Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med1986; 314:1547–1552.
Mancia G, De Backer G, Dominiczak A, et al; ESH-ESC Task Force on the Management of Arterial Hypertension. 2007 ESH-ESC practice guidelines for the management of arterial hypertension: ESH-ESC Task Force on the Management of Arterial Hypertension. J Hypertens2007; 25:1751–1762.
Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. JAMA2003; 289:2560–2572.
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 Med2008; 359:2417–2428.
Chapman N, Chang CL, Dahlöf B, Sever PS, Wedel H, Poulter NR; ASCOT Investigators. Effect of doxazosin gastrointestinal therapeutic system as third-line antihypertensive therapy on blood pressure and lipids in the Anglo-Scandinavian Cardiac Outcomes Trial. Circulation2008; 118:42–48.
de Alvaro F, Hernandez-Presa MAASOCIA Study. Effect of doxazosin gastrointestinal therapeutic system on patients with uncontrolled hypertension: the ASOCIA Study. J Cardiovasc Pharmacol2006; 47:271–276.
Black HR. Doxazosin as combination therapy for patients with stage 1 and stage 2 hypertension. J Cardiovasc Pharmacol2003; 41:866–869.
Chapman N, Chen C-Y, Fujita T, et al. Time to re-appraise the role of alpha-1 adrenoceptor antagonists in the management of hypertension?J Hypertens2010; 28:1796–1803.
Steers WD, Kirby RS. Clinical ease of using doxazosin in BPH patients with and without hypertension. Prostate Cancer Prostatic Dis2005; 8:152–157.
Guthrie RM, Siegel RL. A multicenter, community-based study of doxazosin in the treatment of concomitant hypertension and symptomatic benign prostatic hyperplasia: the Hypertension and BPH Intervention Trial (HABIT). Clin Ther1999; 21:1732–1748.
MacDonald R, Wilt TJ, Howe RW. Doxazosin for treating lower urinary tract symptoms compatible with benign prostatic obstruction: a systematic review of efficacy and adverse effects. BJU Int2004; 94:1263–1270.
Giacomo Rossitto, MD Department of Clinical and Experimental Medicine, University of Padua School of Medicine, Padua, Italy
Ganesh Kamath, MD Department of Medicine, St. Luke’s-Roosevelt Hospital Center, New York, NY
Franz H. Messerli, MD, FACC, FACP Director, Hypertension Program, Professor of Clinical Medicine, Columbia University College of Physicians and Surgeons; Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, New York, NY
Address: Franz H. Messerli, MD, Hypertension Program, Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, 1000 Tenth Avenue, New York, NY 10019; e-mail [email protected]
Giacomo Rossitto, MD Department of Clinical and Experimental Medicine, University of Padua School of Medicine, Padua, Italy
Ganesh Kamath, MD Department of Medicine, St. Luke’s-Roosevelt Hospital Center, New York, NY
Franz H. Messerli, MD, FACC, FACP Director, Hypertension Program, Professor of Clinical Medicine, Columbia University College of Physicians and Surgeons; Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, New York, NY
Address: Franz H. Messerli, MD, Hypertension Program, Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, 1000 Tenth Avenue, New York, NY 10019; e-mail [email protected]
Author and Disclosure Information
Giacomo Rossitto, MD Department of Clinical and Experimental Medicine, University of Padua School of Medicine, Padua, Italy
Ganesh Kamath, MD Department of Medicine, St. Luke’s-Roosevelt Hospital Center, New York, NY
Franz H. Messerli, MD, FACC, FACP Director, Hypertension Program, Professor of Clinical Medicine, Columbia University College of Physicians and Surgeons; Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, New York, NY
Address: Franz H. Messerli, MD, Hypertension Program, Division of Cardiology, St. Luke’s-Roosevelt Hospital Center, 1000 Tenth Avenue, New York, NY 10019; e-mail [email protected]
Alpha-blockers should not be used as first-line therapy for hypertension. However, an alpha-blocker can be considered as a second-line or third-line add-on in a patient whose blood pressure is not under control despite treatment with other drugs.
In addition, alpha-blockers are useful in relieving lower urinary tract symptoms in patients with benign prostatic hypertrophy. However, even in a patient who has both hypertension and benign prostatic hypertrophy, we advise physicians to use alpha-blockers primarily to relieve the urinary symptoms, and we recommend lowering the blood pressure with a drug of a class shown to reduce rates of illness and death.
NOT FIRST-LINE THERAPY
All antihypertensive drugs, including alpha-blockers, lower blood pressure. Alpha-blockers have been approved by the US Food and Drug Administration for treating high blood pressure, and they are just as effective as other antihypertensive drugs—if efficacy is defined as a decrease in millimeters of mercury.
However, lowering the blood pressure is not the main goal of antihypertensive therapy. What we want to achieve when prescribing antihypertensive drugs is to reduce the rates of heart attacks, strokes, and other adverse cardiovascular adverse outcomes, including death.
Unfortunately, alpha-blockers fall short in this regard. In the Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack (ALLHAT) trial,1,2 doxazosin (Cardura) was found to carry a higher risk of combined cardiovascular disease (relative risk 1.19, P = .04), mostly stroke. Alarmingly, the incidence of symptomatic heart failure in patients on doxazosin was twice that in patients on chlorthalidone (relative risk 2.04, P < .001). Doxazosin was minimally more effective in lowering blood pressure than chlorthalidone, but the small difference in blood pressure was unlikely to have accounted for the significant difference in the risk of heart failure.3
This experience with doxazosin illustrates a key drawback to surrogate end points: a treatment may produce a favorable outcome in the surrogate end point (blood pressure) but produce little or no benefit in terms of the real end point (stroke, myocardial infarction, and heart failure).4
Based on the ALLHAT data as well as on a Veterans Administration study in patients with chronic heart failure in which survival with prazosin (Minipress) was no better than with placebo,5 it seems reasonable to no longer use alpha-blockers as initial therapy for hypertension. This view is reflected by current European6 and American7 guidelines.
ALPHA-BLOCKERS AS PART OF COMBINATION THERAPY
In several clinical trials, alpha-blockers were allowed8 or were specified9,10 as add-on therapy if other drugs failed to control the blood pressure, but they were not used in a randomized fashion. Thus, we cannot judge their effect on cardiovascular outcomes such as heart attack and stroke.
The choice of drugs for combination therapy very often is still empirical and based on personal preference. Doxazosin as add-on therapy, in general, has been shown to be safe and well tolerated.11 But even if it is acceptable, it is not a preferred combination.
In the Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT),9 patients received extended-release doxazosin as a third drug if they did not reach their goal blood pressure with either the combination of amlodipine (Norvasc) plus perindopril (Aceon) or atenolol (Tenormin) plus bendroflumethiazide. Extended-release doxazosin was an effective add-on, and there was no apparent excess rate of heart failure in doxazosin users.
In other studies, in patients with uncontrolled hypertension, adding doxazosin as a second- or third-line agent to a gold-standard drug—calcium channel blocker, diuretic, beta-blocker, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, or combinations of these—allowed significantly more participants to achieve their blood pressure goal.11
Personally, we consider doxazosin in patients whose blood pressure is not controlled with triple therapy with a renin-angiotensin system blocker, a diuretic, and a calcium channel antagonist in full doses. In patients with stage 3 or stage 4 kidney disease who can no longer tolerate renin-angiotensin system blockers, doxazosin may also be a useful adjunct. Whether the metabolic effects of alpha-blockers, such as a reduction in insulin resistance and a decrease in total and low-density lipoprotein cholesterol, will result in lower rates of morbidity and death has not been conclusively determined.
A point of view somewhat more favorable to the use of alpha-blockers has recently been put forward by Chapman et al.12
ALPHA-BLOCKERS ALLEVIATE SYMPTOMS OF BENIGN PROSTATIC HYPERTROPHY
Doxazosin and other alpha-blockers are commonly used to alleviate lower urinary tract symptoms in patients with benign prostatic hypertrophy.
Both high blood pressure and benign prostatic hypertrophy become more common with advancing age, and it has been estimated that both are present in more than 25% of men over age 60.13 Indeed, two trials documented that a significant reduction in symptoms of benign prostatic hypertrophy and in systolic and diastolic blood pressure can be achieved with an alpha-blocker.13,14
This raises the question whether such a “twofer” (treating two disease states with one drug) should be used in clinical practice. We have to consider that the principle of the twofer has never been tested and agree with Davis et al,3 who, in a further analysis of the ALLHAT data, stated that, “In older men with benign prostatic hypertrophy in whom an [alpha]-adrenergic blocker seems like the best treatment for the uropathy, coexisting hypertension should be treated with another antihypertensive drug as well.”3
Again, this would clearly relegate doxazosin to second-line or third-line status, even in patients with benign prostatic hypertrophy, in whom it has been shown to be indicated.
ADVERSE EFFECTS OF ALPHA-BLOCKERS
Dizziness, fatigue, and somnolence are occasionally reported but appear to be well tolerated. Postural hypotension is much less common with proper titration of standard doxazosin or with the use of controlled-release formulations.9–15 However, in patients with impaired autonomic function, even long-acting alpha-blockers can cause postural hypotension and syncope.
Patients using phosphodiesterase type 5 inhibitors—sildenafil (Viagra), vardenafil (Levitra), or tadalafil (Cialis)—for erectile dysfunction should avoid alpha-blockers because the blood-pressure-lowering effects of the two drug classes may be additive.
Alpha-blockers should not be used as first-line therapy for hypertension. However, an alpha-blocker can be considered as a second-line or third-line add-on in a patient whose blood pressure is not under control despite treatment with other drugs.
In addition, alpha-blockers are useful in relieving lower urinary tract symptoms in patients with benign prostatic hypertrophy. However, even in a patient who has both hypertension and benign prostatic hypertrophy, we advise physicians to use alpha-blockers primarily to relieve the urinary symptoms, and we recommend lowering the blood pressure with a drug of a class shown to reduce rates of illness and death.
NOT FIRST-LINE THERAPY
All antihypertensive drugs, including alpha-blockers, lower blood pressure. Alpha-blockers have been approved by the US Food and Drug Administration for treating high blood pressure, and they are just as effective as other antihypertensive drugs—if efficacy is defined as a decrease in millimeters of mercury.
However, lowering the blood pressure is not the main goal of antihypertensive therapy. What we want to achieve when prescribing antihypertensive drugs is to reduce the rates of heart attacks, strokes, and other adverse cardiovascular adverse outcomes, including death.
Unfortunately, alpha-blockers fall short in this regard. In the Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack (ALLHAT) trial,1,2 doxazosin (Cardura) was found to carry a higher risk of combined cardiovascular disease (relative risk 1.19, P = .04), mostly stroke. Alarmingly, the incidence of symptomatic heart failure in patients on doxazosin was twice that in patients on chlorthalidone (relative risk 2.04, P < .001). Doxazosin was minimally more effective in lowering blood pressure than chlorthalidone, but the small difference in blood pressure was unlikely to have accounted for the significant difference in the risk of heart failure.3
This experience with doxazosin illustrates a key drawback to surrogate end points: a treatment may produce a favorable outcome in the surrogate end point (blood pressure) but produce little or no benefit in terms of the real end point (stroke, myocardial infarction, and heart failure).4
Based on the ALLHAT data as well as on a Veterans Administration study in patients with chronic heart failure in which survival with prazosin (Minipress) was no better than with placebo,5 it seems reasonable to no longer use alpha-blockers as initial therapy for hypertension. This view is reflected by current European6 and American7 guidelines.
ALPHA-BLOCKERS AS PART OF COMBINATION THERAPY
In several clinical trials, alpha-blockers were allowed8 or were specified9,10 as add-on therapy if other drugs failed to control the blood pressure, but they were not used in a randomized fashion. Thus, we cannot judge their effect on cardiovascular outcomes such as heart attack and stroke.
The choice of drugs for combination therapy very often is still empirical and based on personal preference. Doxazosin as add-on therapy, in general, has been shown to be safe and well tolerated.11 But even if it is acceptable, it is not a preferred combination.
In the Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT),9 patients received extended-release doxazosin as a third drug if they did not reach their goal blood pressure with either the combination of amlodipine (Norvasc) plus perindopril (Aceon) or atenolol (Tenormin) plus bendroflumethiazide. Extended-release doxazosin was an effective add-on, and there was no apparent excess rate of heart failure in doxazosin users.
In other studies, in patients with uncontrolled hypertension, adding doxazosin as a second- or third-line agent to a gold-standard drug—calcium channel blocker, diuretic, beta-blocker, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, or combinations of these—allowed significantly more participants to achieve their blood pressure goal.11
Personally, we consider doxazosin in patients whose blood pressure is not controlled with triple therapy with a renin-angiotensin system blocker, a diuretic, and a calcium channel antagonist in full doses. In patients with stage 3 or stage 4 kidney disease who can no longer tolerate renin-angiotensin system blockers, doxazosin may also be a useful adjunct. Whether the metabolic effects of alpha-blockers, such as a reduction in insulin resistance and a decrease in total and low-density lipoprotein cholesterol, will result in lower rates of morbidity and death has not been conclusively determined.
A point of view somewhat more favorable to the use of alpha-blockers has recently been put forward by Chapman et al.12
ALPHA-BLOCKERS ALLEVIATE SYMPTOMS OF BENIGN PROSTATIC HYPERTROPHY
Doxazosin and other alpha-blockers are commonly used to alleviate lower urinary tract symptoms in patients with benign prostatic hypertrophy.
Both high blood pressure and benign prostatic hypertrophy become more common with advancing age, and it has been estimated that both are present in more than 25% of men over age 60.13 Indeed, two trials documented that a significant reduction in symptoms of benign prostatic hypertrophy and in systolic and diastolic blood pressure can be achieved with an alpha-blocker.13,14
This raises the question whether such a “twofer” (treating two disease states with one drug) should be used in clinical practice. We have to consider that the principle of the twofer has never been tested and agree with Davis et al,3 who, in a further analysis of the ALLHAT data, stated that, “In older men with benign prostatic hypertrophy in whom an [alpha]-adrenergic blocker seems like the best treatment for the uropathy, coexisting hypertension should be treated with another antihypertensive drug as well.”3
Again, this would clearly relegate doxazosin to second-line or third-line status, even in patients with benign prostatic hypertrophy, in whom it has been shown to be indicated.
ADVERSE EFFECTS OF ALPHA-BLOCKERS
Dizziness, fatigue, and somnolence are occasionally reported but appear to be well tolerated. Postural hypotension is much less common with proper titration of standard doxazosin or with the use of controlled-release formulations.9–15 However, in patients with impaired autonomic function, even long-acting alpha-blockers can cause postural hypotension and syncope.
Patients using phosphodiesterase type 5 inhibitors—sildenafil (Viagra), vardenafil (Levitra), or tadalafil (Cialis)—for erectile dysfunction should avoid alpha-blockers because the blood-pressure-lowering effects of the two drug classes may be additive.
References
Messerli FH. Implications of discontinuation of doxazosin arm of ALLHAT. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (commentary). Lancet2000; 355:863–864.
Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). ALLHAT Collaborative Research Group. JAMA2000; 283:1967–1975.
Davis BR, Cutler JA, Furberg CD, et al; ALLHAT Collaborative Research Group. Relationship of antihypertensive treatment regimens and change in blood pressure to risk for heart failure in hypertensive patients randomly assigned to doxazosin or chlorthalidone: further analyses from the Antihypertensive and Lipid-Lowering treatment to prevent Heart Attack Trial. Ann Intern Med2002; 137:313–320.
Messerli FH. Doxazosin and congestive heart failure (viewpoint). J Am Coll Cardiol2001; 38:1295–1296.
Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med1986; 314:1547–1552.
Mancia G, De Backer G, Dominiczak A, et al; ESH-ESC Task Force on the Management of Arterial Hypertension. 2007 ESH-ESC practice guidelines for the management of arterial hypertension: ESH-ESC Task Force on the Management of Arterial Hypertension. J Hypertens2007; 25:1751–1762.
Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. JAMA2003; 289:2560–2572.
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 Med2008; 359:2417–2428.
Chapman N, Chang CL, Dahlöf B, Sever PS, Wedel H, Poulter NR; ASCOT Investigators. Effect of doxazosin gastrointestinal therapeutic system as third-line antihypertensive therapy on blood pressure and lipids in the Anglo-Scandinavian Cardiac Outcomes Trial. Circulation2008; 118:42–48.
de Alvaro F, Hernandez-Presa MAASOCIA Study. Effect of doxazosin gastrointestinal therapeutic system on patients with uncontrolled hypertension: the ASOCIA Study. J Cardiovasc Pharmacol2006; 47:271–276.
Black HR. Doxazosin as combination therapy for patients with stage 1 and stage 2 hypertension. J Cardiovasc Pharmacol2003; 41:866–869.
Chapman N, Chen C-Y, Fujita T, et al. Time to re-appraise the role of alpha-1 adrenoceptor antagonists in the management of hypertension?J Hypertens2010; 28:1796–1803.
Steers WD, Kirby RS. Clinical ease of using doxazosin in BPH patients with and without hypertension. Prostate Cancer Prostatic Dis2005; 8:152–157.
Guthrie RM, Siegel RL. A multicenter, community-based study of doxazosin in the treatment of concomitant hypertension and symptomatic benign prostatic hyperplasia: the Hypertension and BPH Intervention Trial (HABIT). Clin Ther1999; 21:1732–1748.
MacDonald R, Wilt TJ, Howe RW. Doxazosin for treating lower urinary tract symptoms compatible with benign prostatic obstruction: a systematic review of efficacy and adverse effects. BJU Int2004; 94:1263–1270.
References
Messerli FH. Implications of discontinuation of doxazosin arm of ALLHAT. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (commentary). Lancet2000; 355:863–864.
Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). ALLHAT Collaborative Research Group. JAMA2000; 283:1967–1975.
Davis BR, Cutler JA, Furberg CD, et al; ALLHAT Collaborative Research Group. Relationship of antihypertensive treatment regimens and change in blood pressure to risk for heart failure in hypertensive patients randomly assigned to doxazosin or chlorthalidone: further analyses from the Antihypertensive and Lipid-Lowering treatment to prevent Heart Attack Trial. Ann Intern Med2002; 137:313–320.
Messerli FH. Doxazosin and congestive heart failure (viewpoint). J Am Coll Cardiol2001; 38:1295–1296.
Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med1986; 314:1547–1552.
Mancia G, De Backer G, Dominiczak A, et al; ESH-ESC Task Force on the Management of Arterial Hypertension. 2007 ESH-ESC practice guidelines for the management of arterial hypertension: ESH-ESC Task Force on the Management of Arterial Hypertension. J Hypertens2007; 25:1751–1762.
Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. JAMA2003; 289:2560–2572.
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 Med2008; 359:2417–2428.
Chapman N, Chang CL, Dahlöf B, Sever PS, Wedel H, Poulter NR; ASCOT Investigators. Effect of doxazosin gastrointestinal therapeutic system as third-line antihypertensive therapy on blood pressure and lipids in the Anglo-Scandinavian Cardiac Outcomes Trial. Circulation2008; 118:42–48.
de Alvaro F, Hernandez-Presa MAASOCIA Study. Effect of doxazosin gastrointestinal therapeutic system on patients with uncontrolled hypertension: the ASOCIA Study. J Cardiovasc Pharmacol2006; 47:271–276.
Black HR. Doxazosin as combination therapy for patients with stage 1 and stage 2 hypertension. J Cardiovasc Pharmacol2003; 41:866–869.
Chapman N, Chen C-Y, Fujita T, et al. Time to re-appraise the role of alpha-1 adrenoceptor antagonists in the management of hypertension?J Hypertens2010; 28:1796–1803.
Steers WD, Kirby RS. Clinical ease of using doxazosin in BPH patients with and without hypertension. Prostate Cancer Prostatic Dis2005; 8:152–157.
Guthrie RM, Siegel RL. A multicenter, community-based study of doxazosin in the treatment of concomitant hypertension and symptomatic benign prostatic hyperplasia: the Hypertension and BPH Intervention Trial (HABIT). Clin Ther1999; 21:1732–1748.
MacDonald R, Wilt TJ, Howe RW. Doxazosin for treating lower urinary tract symptoms compatible with benign prostatic obstruction: a systematic review of efficacy and adverse effects. BJU Int2004; 94:1263–1270.
Monoamine oxidase (MAO) inhibitors were the first drugs for treating depression. Introduced in the 1950s, they were used extensively for the next two decades. Their use declined substantially since then because of their reported side effects, their food and drug interactions, and the introduction of new classes of antidepressants.
This trend may be changing. These drugs can be effective in major depressive disorder, and particularly in major depressive disorder with atypical features and in treatment-resistant depression.
New, selective MAO inhibitors are being developed. Moreover, the selegiline transdermal system (Emsam),1,2 introduced in 2006, offers the potential advantage of eliminating the need for burdensome dietary restrictions and has renewed interest in this group of drugs.
In this article, we discuss the history, pharmacology, safety and tolerability of MAO inhibitors, and we summarize recent MAO inhibitor research. Our goal is to familiarize physicians with this class of drugs, including recent updates regarding their safety profile and liberalized dietary recommendations.
DEPRESSION IS COMMON, DIFFICULT
Depression affects 121 million people worldwide.3 According to a study that compared two surveys of 40,000 people each, the prevalence of major depressive disorder in the United States more than doubled (from 3.3% to 7.0%) from 1992 to 2002.4 Another survey, in 2002 and 2003, revealed the lifetime prevalence of major depressive disorder to be 16.6%.5
Treatments for depression have expanded over the past 20 years, with new classes of drugs such as selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs). However, depression has remained a difficult condition to treat. In the National Institute of Mental Health’s Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study,6 the remission rate in patients treated with the SSRI citalopram (Celexa) for up to 14 weeks was 28% using one measure and 33% using another. Diversifying and understanding existing and emerging therapeutic options is important to the effective treatment of this disease.
THE RISE AND FALL OF MAO INHIBITORS
The first antidepressant introduced was an MAO inhibitor, iproniazid, followed shortly thereafter by a tricyclic antidepressant, imipramine (Tofranil). When iproniazid, originally an antituberculosis agent, was promoted for its antidepressant properties in the 1950s, very little was known about its side effects. It was later removed from the market because of hepatotoxicity, but several other MAO inhibitors had surfaced for the treatment of depression—eg, phenelzine (Nardil), isocarboxazid (Marplan), and tranylcypromine (Parnate).
Currently, MAO inhibitors are typically reserved for third- or fourth-line treatment. As a result, even psychiatrists have little experience with these agents. In a 1999 survey of the Michigan Psychiatric Association,7 12% of practicing psychiatrists said they had never prescribed an MAO inhibitor, another 27% had not prescribed one in the prior 3 years, and only 2% said they prescribed them frequently. A decade earlier, about 25% had said they prescribed them often.8
The prescription rate of MAO inhibitors has remained low during the past 10 years. In a Canadian population-based study9 conducted among older adults in a large health care database from January 1997 to April 2007, the yearly incidence of MAO inhibitor prescriptions decreased from a rate of 3.1 per 100,000 to 1.4 per 100,000. Drug interactions, side effects, preference for other treatments, and dietary restrictions were the reasons most often cited for not prescribing these drugs.7
The side effects of MAO inhibitors were recognized by the mid-1960s, when more than 40 cases of tyramine-induced hypertensive crisis were reported (particularly with tranylcypromine).10,11 Many of the reported events happened after the patient ate tyramine-rich foods such as aged cheese (hence, “the cheese reaction”—more on this below) or drank draft beer.10,11 The US Food and Drug Administration (FDA) consequently established dietary restrictions for patients taking MAO inhibitors, but people found the guidelines cumbersome and often switched to newer drugs that did not require a restrictive diet, such as tricyclics and, much later (in the 1980s), SSRIs.
MAO HAS TWO SUBTYPES
MAO is a flavin-containing enzyme critical for regulating neurotransmitter levels by catabolizing endogenous monoamines (eg, norepinephrine, serotonin, and dopamine) and exogenous amines (eg, dietary tyramine). It is found throughout the body but is more highly concentrated in the liver, kidneys, intestinal wall, and brain.
MAO has two subtypes, isoenzyme A (MAO-A) and isoenzyme B (MAO-B), which vary in their distribution. MAO-A is found primarily in the intestinal tract, liver, and peripheral adrenergic neurons (adrenal glands, arterial vessels, and sympathetic nerves) and preferentially metabolizes serotonin and norepinephrine. MAO-B is found mostly in the brain and liver. However, both isotypes are found in all of the areas mentioned. Since 80% of intestinal MAO is MAO-A, this isoenzyme is primarily responsible for degradation of tyramine, and thus inhibition of MAO-A is associated with the cheese reaction.10,11
TYPES OF MAO INHIBITORS
MAO inhibitors can be classified on the basis of whether they are nonselective or selective for either MAO-A or MAO-B, and whether their effect is reversible.
Nonselective MAO inhibitors are phenelzine, isocarboxazid, and tranylcypromine.
Selective MAO inhibitors. Selegiline is selective for MAO-B. Clorgyline is selective for MAO-A, but it is not available in the United States.
A reversible MAO inhibitor is moclobemide (not available in the United States).
Do selectivity and reversibility matter?
Classic MAO inhibitors such as tranylcypromine and phenelzine are neither reversible (binding to the enzyme for the extent of its lifetime of 14–28 days) nor selective for the subtypes. These drugs were used extensively several decades ago to treat atypical depression, anxiety, and phobias. The only selective MAO inhibitor now available in the United States is selegiline, which inhibits MAO-B at low doses but loses its selectivity at dosages greater than 20 mg/day.
Experimental studies suggest that inhibition of more than 70% of MAO-A activity is necessary for the antidepressant effect of selegiline.12 At oral doses that selectively inhibit MAO-B (5–10 mg/day), selegiline does not seem to have potent antidepressant activity, although it does show success as an adjunctive treatment for Parkinson disease and does not necessitate any dietary restriction. Only at higher oral doses (20–60 mg/day), at which MAO-B selectivity is lost, is the antidepressant effect seen. But the higher doses necessitate dietary restrictions. Therefore, patients who are taking the oral selective MAO inhibitor selegiline have to follow the same dietary restrictions as patients taking the nonselective ones.
Reversible inhibitors of MAO-A have the distinction of being easily displaced by ingested tyramine in the gut and thus do not cause the cheese reaction. However, the only reversible agent available in the world market is moclobemide. It is not available in the United States, and appears to be less effective than older, nonselective MAO inhibitors.13
SELEGILINE TRANSDERMAL SYSTEM
The selegiline transdermal system (Emsam) is the first FDA-approved transdermal patch for treatment of major depression. Patients who are using Emsam at its lowest effective dose of 6 mg/24 hours do not need to follow the dietary restrictions that are needed for all oral MAO inhibitors.
Pharmacokinetics of the selegiline patch
With the transdermal patch, selegiline is extensively absorbed through the skin. Plasma levels are maintained over a 24-hour period, allowing once-daily application. Patches are available that deliver 6, 9, or 12 mg per 24 hours. Steady-state plasma levels are reached after about 5 days.
The bioavailability of selegiline is about 75% with the transdermal delivery system vs 4.4% after oral administration, the lower number being due to first-pass metabolism.1 About 90% of selegiline is bound to plasma proteins and quickly penetrates the central nervous system.
This drug is metabolized by cytochrome P450 isoenzymes, including CYP2C9, CYP2B6, and CYP3A4. Its metabolites are l-methamphetamine and n-desmethylselegiline.
Clinical research showed that dosage adjustments were not necessary in specific populations studied, including patients with various stages of renal or hepatic failure.1 Clearance of selegiline was independent of dose, age, sex, renal function, body weight, or concomitant medications.1
Advantages of the patch system
Since selegiline delivered via the patch is not absorbed through the gut, it has little effect on gut MAO-A and therefore is unlikely to lead to tyramine-induced hypertensive crisis. Studies of the selegiline patch show that inhibition of more than 80% of gut MAO-A is necessary to impair metabolism of tyramine in the gut.1 Therefore, the 6-mg patch will not significantly impair tyramine degradation in the gut. In phase III testing of the selegiline patch, no hypertensive crises were reported among 2,656 outpatients without dietary restrictions. However, it is still recommended that patients on the 9-mg and 12-mg patches follow a tyramine-free diet.1
Although there are no data available to suggest that higher dosages are more effective, it is recommended that the dose be titrated in 3-mg increments at intervals of at least 2 weeks until the maximum recommended dosage of 12 mg/24 hours is reached.2
Disadvantage of the selegiline patch: Cost
The selegiline patch is expensive: $692.99 for 1 month’s supply at a dose of 6 mg/24 hours and $638.99 for 1 month’s supply at a dose of 9 or 12 mg/24 hours (verified with a national pharmacy chain at the time of this writing). Insurance coverage for the patch varies, and documentation may be required from the physician. Oral MAO inhibitors are much less expensive.
SAFETY, TOLERABILITY OF MAO INHIBITORS
Side effects of oral agents
Orthostatic hypotension, dizziness, drowsiness, insomnia, and nausea are the most frequently reported side effects of oral MAO inhibitors.14,15 These side effects can generally be managed symptomatically by slowing the titration, dividing the doses, changing the time it is taken, or, in the case of orthostatic hypotension, increasing fluid intake.14 Phenelzine has the strongest association with sedation.14
Weight gain, edema, muscle pain, myoclonus, paresthesias, sexual dysfunction, and, rarely, hepatotoxicity are late side effects.15–18 Paresthesias, an infrequent side effect, are often treated with pyridoxine supplementation.15
Transient hypertensive episodes within 2 hours after ingestion of MAO inhibitors, which were independent of dietary or drug interactions, have been reported.19 The hypertensive episodes are usually self-limited but in rare cases result in hypertensive crisis.19–21
Serotonin syndrome has been reported with MAO inhibitor monotherapy in rare cases.22 Serotonin syndrome is characterized by mental status changes, restlessness, myoclonus, hyperreflexia, diaphoresis, or evidence of autonomic hyperactivity.23 The syndrome is potentially fatal and is treated symptomatically by removing the offending drugs and giving intravenous rehydration.23
Side effects of the selegiline patch
The most common adverse events with the selegiline patch include application-site reaction (24% vs 12% with placebo), headache (18% vs 17%), insomnia, diarrhea, dry mouth, and dyspepsia.24,25 Dose-related orthostatic hypotension was reported (occurring in 9.8% vs 6.7% with placebo) and was most likely to occur in elderly patients.25 It is suggested that insomnia may be lessened by removing the patch before bedtime. Also, rotating the patch application sites and prompt topical treatment of irritation may lessen local effects.24
Observe a washout period when switching between serotonergic drugs
Most MAO inhibitors irreversibly inhibit MAO for the life of the enzyme, and thus the physiologic effects of phenelzine, isocarboxazid, and tranylcypromine last for up to 2 to 3 weeks.26 Although the elimination half-life of typical MAO inhibitors is short (1.5–4 hours),27,28 their physiologic effects are long-lasting.14
Switching from a MAO inhibitor to another serotonergic agent. Concomitant use of MAO inhibitors and other serotonergic drugs is associated with the risk of serotonin syndrome. After stopping an MAO inhibitor, a 14-day washout period is recommended before starting another serotonergic agent.29 Patients should continue to be monitored closely after the washout period, as cases of serotonin syndrome have been reported later.30 A 14-day washout period is also recommended when switching between MAO inhibitors, although more rapid switches have been made safely.31
Switching from another serotonergic agent to an oral MAO inhibitor. Similarly, a 14-day washout period (or five half-lives) is necessary after stopping most of the serotonergic agents mentioned above before beginning treatment with an oral MAO inhibitor. Fluoxetine (Prozac) has a longer half-life and therefore requires a longer washout period, ie, 5 weeks.
Switching from another serotonergic agent to the selegiline patch. When switching to the selegiline patch from another serotonergic drug, the washout period is 1 week after stopping most drugs or 5 weeks after stopping fluoxetine. One must wait 2 weeks after stopping the selegiline patch before starting therapy with any of the other serotonergic drugs.
Drugs to avoid due to interactions
In view of the risk of severe of drug-drug interactions, particularly the risk of serotonin syndrome, the following serotonin-enhancing compounds are contraindicated in patients taking a MAO inhibitor: SSRIs, SNRIs, tricyclic antidepressants, other MAO inhibitors, mirtazapine, and St. John’s wort. Other pharmaceuticals to be avoided include bupropion, meperidine, tramadol, methadone, propoxyphene, pentazocine, dextromethorphan, and cyclobenzaprine (Table 1). Also, there have been numerous reports of serotonin syndrome with the use of the broad-spectrum, MAO-based antibiotic linezolid (Zyvox), by itself or in conjunction with other serotonergic agents.32–35
Several studies suggested a hazardous combination of nonsubcutaneous sumatriptans (5-HT1B/1D agonists) and MAO-B inhibitors, while subcutaneous sumatriptan migraine-abortive treatment and MAO-B inhibitors appear to be safe.36,37
Also, amphetamines, cough-and-cold preparations, and weight-reducing preparations that contain vasoconstrictors (eg, pseudoephedrine, phenylephrine, phenylpropanolamine, and ephedrine) should be avoided, as the risk of hypertensive crisis increases with these products.
Patients on MAO inhibitors should wear a medical alert bracelet in case they need to undergo emergency surgery and are unable to verbally communicate their drug history. They should be instructed to alert all health care providers about their MAO inhibitor use.14
Beware of worsening depression
Physicians, patients, and family members should be advised to observe for worsening depression or “suicidality” during the course of treatment with MAO inhibitors, as with all antidepressants.
Diet can be more lenient than in the past
The dietary restrictions classically advised for patients taking oral MAO inhibitors were established to prevent hypertensive crises associated with tyramine ingestion. However, some of these restrictions were unsubstantiated,38 and evidence from more recent studies suggests that they are unnecessarily strict39 and may lead to resistance by the physician, the patient, or both to using this potentially beneficial therapy.14 There is also a risk that patients will inadvertently discover that a food that was in the “restricted” list caused them no harm upon ingestion and thus will become cavalier about dietary adherence.39
To prevent dietary noncompliance, physicians should conduct ongoing diet surveys and encourage adherence to evidence-based dietary recommendations.40
The FDA and drug-package inserts for oral MAO inhibitors continue to recommend stringent dietary restrictions, including no aged cheeses or meats, soy sauce, soy beans, soy paste, miso soup, Italian green beans (fava beans), snow peas, broad bean pods, sauerkraut, kimchee, concentrated yeast extracts (Marmite), wine, beer (including alcohol-free beer), and many other foods. However, several studies have measured the tyramine content of food and determined that less than 6 mg per serving is generally safe.39,41 The results of these investigations have led to more lenient dietary guidelines.39
Absolute dietary restrictions include39:
Aged cheeses and meats
Banana peels
Broad bean (fava) pods
Spoiled meats
Marmite
Sauerkraut
Soybean products
Draft beers.
Among the many foods determined to be unnecessarily restricted are avocados; bananas; beef or chicken bouillon; chocolate; fresh and mild cheeses, eg, ricotta, cottage cheese, cream cheese, processed cheese slices; fresh meat, poultry, or fish; meat gravy (fresh); monosodium glutamate; peanuts; properly stored pickled or smoked fish (eg, herring); raspberries; and yeast extracts (except Marmite).39
Dietary restrictions should continue for 2 weeks after stopping an MAO inhibitor.
Dietary restrictions for the selegiline patch
Tyramine-containing foods pose less risk with the selegiline patch than with oral MAO inhibitors, and studies42 show that the 6-mg patch does not necessitate dietary restrictions. The accumulating data suggest that the risk of a tyramine-induced event is extremely low with the patch even in doses above 6 mg. But in the meantime, the recommendations for the 9-mg and 12-mg patches remain the same as for the classic oral MAO inhibitors, and tyramine-containing food should be restricted.
EFFICACY OF MAO INHIBITORS IN CLINICAL PRACTICE
Data from numerous studies suggest MAO inhibitors are effective in managing major depressive disorder, and specifically atypical depression,43–48 treatment-resistant major depressive disorder,49,50 and bipolar depression.51,52 Guidelines from the American Psychiatric Association and the British Association for Psychopharmacology suggest that MAO inhibitors be recommended for treatment of major depressive disorder in patients with atypical features and when other antidepressants have failed.53
MAO inhibitors have also been used in the treatment of Parkinson disease, bulimia, anxiety disorders, anorexia nervosa, and body dysmorphic disorder.54
Major depressive disorder
In controlled trials in outpatients with depression who were treated with therapeutic doses of MAO inhibitors, the response rate was 50% to 70%.55 When tranylcypromine was used in severely depressed inpatients, its efficacy was comparable to that of electroconvulsive therapy, imipramine, and amitriptyline.56 Thase et al,57 in a meta-analysis, found that the MAO inhibitors tranylcypromine, phenelzine, and isocarboxazid were equally effective in treating depression.
Atypical depression
Atypical depression is one of the most common subtypes of major depressive disorder. Diagnostic criteria for major depressive disorder with atypical features include mood reactivity and two of the following: weight gain or hyperphagia, hypersomnolence, leaden paralysis, or an enduring pattern of rejection sensitivity.58 An estimated 30% of outpatients with unipolar depression meet these criteria.59
Multiple randomized controlled trials showed that MAO inhibitors were superior to tricyclic antidepressants in treating atypical depression. One study, involving more than 400 patients, determined that atypical depression responded better to phenelzine than to imipramine.43 Another study evaluating 153 critically depressed patients showed significantly greater response with phenelzine than with imipramine or placebo.49 Furthermore, in another double-blind controlled crossover study, 89 mood-reactive, nonmelancholic, chronically depressed outpatients were found to have a striking response to phenelzine after being unresponsive to imipramine.50 Another report48 indicated that in a double-blind, randomized, placebo-controlled trial among 119 patients with atypical depression treated for 6 weeks, the overall response rates were 78% with phenelzine, 50% with imipramine, and 28% with placebo.
A recent meta-analysis of treatment trials in atypical depression revealed a large mean effect size of 0.45 for the superiority of MAO inhibitors over placebo and a medium mean effect size of 0.27 for the superiority of MAO inhibitors over tricyclic antidepressants.60 Additionally, in a randomized, double-blind placebo-controlled trial, patients with comorbid atypical depression and bulimia showed significant improvement in both bulimic and depressive symptoms when given phenelzine vs imipramine or placebo.61
The current data comparing SSRIs and MAO inhibitors in the treatment of atypical depression are limited. The above-mentioned meta-analysis of three such trials (when moclobemide was used in two out of three trials) revealed no significant difference in efficacy.60 However, the authors themselves warned about the limitations of the studies, including low power to detect differences. Parker and Crawford59 compared self-rating of effectiveness of the various previous treatments in patients with depression with and without atypical features using an online survey. The analysis of the responses of 1,934 patients showed no overall difference in treatment response to both drug and non-drug therapies between respondents with and without atypical features, except with SSRIs. The “atypical” group had a significantly lower mean effectiveness score for SSRIs overall, and a lower mean effectiveness rating for two of six SSRIs examined. The authors speculated that even though there was no differential outcome detected in individuals with atypical depression treated with MAO inhibitors, this negative finding may simply have reflected the low prevalence of sample respondents who received MAO inhibitors (which was 4% in the “atypical depression” group of 338).59
Treatment-resistant depression
The ultimate goal in treating major depressive disorder is to achieve complete remission. If complete remission is not achieved, the risk of relapse is high,62,63 as is the risk of more severe future depressive episodes63 and death from any cause.64 Therefore, the ability of clinicians to make appropriate and evidence-based changes in treatment strategy is of high importance.
The use of MAO inhibitors as a third-line or fourth-line choice for treatment-resistant depression is supported by a number of studies.49,50,65–67 MAO inhibitors appear to be especially effective in the subgroup of patients who have treatment-resistant depression with atypical or anergic bipolar features.
Bipolar depression
Anergic bipolar depression is defined as a condition associated with fatigue, psychomotor retardation, and at least one reversed neurovegetative symptom in a patient with bipolar disorder meeting the criteria for a major depressive episode. According to several trials,51,52,68 MAO inhibitors may be more effective than a tricyclic antidepressant in the treatment of anergic bipolar depression. However, more studies are required to determine the role of antidepressants in general and MAO inhibitors in particular in the management of bipolar depression.
Efficacy of the selegiline patch
The efficacy of the selegiline patch in the treatment of depression was examined in four double-blind placebo-controlled studies.69–72 There were three short-term studies (a 6-week study of 177 patients,69 an 8-week study of 265 patients,72 and an 8-week study of 289 patients70) and a fixed-dose 1-year relapse prevention study of 322 patients.71 The inclusion criterion for the short-term studies was diagnosis of a first or a recurrent episode of major depressive disorder in patients with a Hamilton Depression Rating Scale (HDRS) score higher than 20. The HDRS score was used to assess improvement in depressive symptoms. In all studies, patients on active patch had significant improvement in depressive symptoms on the HDRS compared with placebo. In the relapse prevention study,71 patients with major depressive disorder that responded to transdermal selegiline 6 mg within the first 10 weeks were stratified either to continue receiving the selegiline 6-mg patch or to receive placebo. Those continually receiving selegiline experienced a significantly longer time to relapse. At 12 months, the relapse rate was 16.8% with the selegiline patch vs 30.7% with placebo. The patch was reported to be well tolerated, with the most common side effect being application site reaction. The adherence to the treatment was high—84.2% in the active-patch group and 89.6% in the placebo group.71
DO MAO INHIBITORS HAVE A PLACE IN PRIMARY CARE?
MAO inhibitors have secured their place in the history of psychiatry as the first antidepressants. Overall, MAO inhibitors remain underused. However, with the introduction of new and selective MAO inhibitors including the selegiline patch, and with data suggesting efficacy in the management of certain subtypes of depression, we expect that interest in this class of drugs will grow among psychiatrists. Based on the current guidelines for MAO inhibitors to be used as a third- or fourth-line treatment, as well as on research data, it is premature to recommend their more extensive use in a primary care setting. Whether this will change in the future depends on both the research advances and new, safer formulations of MAO inhibitors.
Compton WM, Conway KP, Stinson FS, Grant BF. Changes in the prevalence of major depression and comorbid substance use disorders in the United States between 1991–1992 and 2001–2002. Am J Psychiatry2006; 163:2141–2147.
Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry2005; 62:593–602.
Trivedi MH, Rush AJ, Wisniewski SR, et al; STAR*D Study Team. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry2006; 163:28–40.
Balon R, Mufti R, Arfken CL. A survey of prescribing practices for monoamine oxidase inhibitors. Psychiatr Serv1999; 50:945–947.
Clary C, Mandos LA, Schweizer E. Results of a brief survey on the prescribing practices for monoamine oxidase inhibitor antidepressants. J Clin Psychiatry1990; 51:226–231.
Shulman KI, Fischer HD, Herrmann N, Huo CY, Anderson GM, Rochon PA. Current prescription patterns and safety profile of irreversible monoamine oxidase inhibitors: a population-based cohort study of older adults. J Clin Psychiatry2009; 70:1681–1686.
Horwitz D, Lovenberg W, Engelman K, Sjoerdsma A. Monoamine oxidase inhibitors, tyramine, and cheese. JAMA1964; 188:1108–1110.
Asatoor AM, Levi AJ, Milne MD. Tranylcypromine and cheese. Lancet1963; 2:733–734.
Gordon MN, Muller CD, Sherman KA, Morgan DG, Azzaro AJ, Wecker L. Oral versus transdermal selegiline: antidepressant-like activity in rats. Pharmacol Biochem Behav1999; 63:501–506.
Lotufu-Neto F, Trivedi M, Thase ME. Meta-analysis of the reversible inhibitors of monoamine oxidase type A moclobemide and brofaromine for the treatment of depression. Neuropsychopharmacology1999; 20:226–247.
Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract2004; 10:239–248.
Evans DL, Davidson J, Raft D. Early and late side effects of phenelzine. J Clin Psychopharmacol1982; 2:208–210.
Fava M. Weight gain and antidepressants. J Clin Psychiatry2000; 61(suppl 11):37–41.
Rabkin J, Quitkin F, Harrison W, Tricamo E, McGrath P. Adverse reactions to monoamine oxidase inhibitors. Part I. A comparative study. J Clin Psychopharmacol1984; 4:270–278.
Gomez-Gil E, Salmeron JM, Mas A. Phenelzine-induced fulminant hepatic failure. Ann Intern Med1996; 124:692–693.
Lavin MR, Mendelwitz A, Kronig MH. Spontaneous hypertensive reactions with monoamine oxidase inhibitors. Biol Psychiatry1993; 34:146–151.
Fisher P. Serotonin syndrome in the elderly after antidepressive monotherapy. J Clin Psychopharmacol1995; 15:440–442.
Sternback H. The serotonin syndrome. Am J Psychiatry1991; 148:705–713.
Thase M. Novel transdermal delivery formulation of the monoamine oxidase inhibitor selegiline nearing release for treatment of depression. J Clin Psychiatry2006; 67:671–672.
Lee KC, Chen JJ. Transdermal selegiline for the treatment of major depressive disorder. Neuropsychiatr Dis Treat2007; 3:527–537.
Cooper AJ. Tyramine and irreversible monoamine oxidase inhibitors in clinical practice. Br J Psychiatry Suppl1989; Oct(6):38–45.
Mallinger AG, Smith E. Pharmacokinetics of monoamine oxidase inhibitors. Psychopharmacol Bull1991; 27:493–502.
Fulton B, Benfield P. Moclobemide. An update of its pharmacological properties and therapeutic use. Drugs1996; 52:450–474.
Marangell LB. Switching antidepressants for treatment-resistant major depression. J Clin Psychiatry2001; 62(suppl 18):12–17.
Gitlin MJ. Venlafaxine, monoamine oxidase inhibitors, and the serotonin syndrome. J Clin Psychopharmacol1997; 17:66–67.
Szuba MP, Hornig-Rohan M, Amsterdam JD. Rapid conversion from one monoamine oxidase inhibitor to another. J Clin Psychiatry1997; 58:307–310.
Lorenz RA, Vandenberg AM, Canepa EA. Serotonergic antidepressants and linezolid: a retrospective chart review and presentation of cases. Int J Psychiatry Med2008; 38:81–90.
Miller DG, Lovell EO. Antibiotic-induced serotonin syndrome. J Emerg Med2008, May1(Epub ahead of print).
Das PK, Wakentin DI, Hewko R, Forrest DL. Serotonin syndrome after concomitant treatment with linezolid and meperidine. Clin Infect Dis2008; 46:264–265.
Packer S, Berman SA. Serotonin syndrome precipitated by the monoamine oxidase inhibitor linezolid. Am J Psychiatry2007; 164:346–347.
Diamond S. The use of sumatriptan in patients on monoamine oxidase inhibitors. Neurology1995; 45:1039–1040.
Fox AW. Subcutaneous sumatriptan pharmacokinetics: delimiting the monoamine oxidase inhibitor effect. Headache2010; 50:249–255.
Gardner DM, Shulman KI, Walker SE, Tailor SA. The making of a user friendly MAOI diet. J Clin Psychiatry1996; 57:99–104.
Sweet RA, Brown EJ, Heimberg RG, et al. Monoamine oxidase inhibitor dietary restrictions: what are we asking patients to give up?J Clin Psychiatry1995; 56:196–201.
Shulman KI, Walker SE. Refining the MAOI diet: tyramine content of pizzas and soy products. J Clin Psychiatry1999; 60:191–193.
Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol2006; 46:933–944.
Quitkin FM, Stewart JW, McGrath PJ, et al. Columbia atypical depression. A subgroup of depressives with better response to MAOI than to tricyclic antidepressants or placebo. Br J Psychiatry Suppl1993; Sep(21):30–34.
Davidson J, Pelton S. Forms of atypical depression and their response to antidepressant drugs. Psychiatry Res1986; 17:87–95.
Liebowitz MR, Quitkin FM, Stewart JW, et al. Antidepressant specificity in atypical depression. Arch Gen Psychiatry1988; 45:129–137.
Rapaport MH, Thase ME. Translating the evidence on atypical depression into clinical practice. J Clin Psychiatry2007; 68:e11.
Liebowitz MR. Depression with anxiety and atypical depression. J Clin Psychiatry1993; 54(suppl):10–14.
Stewart JW, McGrath PJ, Quitkin FM, et al. Chronic depression: response to placebo, imipramine, and phenelzine. J Clin Psychopharmacol1993; 13:391–396.
McGrath PJ, Stewart JW, Nunes EV, et al. A double-blind crossover trial of imipramine and phenelzine for outpatients with treatment-refractory depression. Am J Psychiatry1993; 150:118–123.
Zarate CA, Tohen M, Baraibar G, Kando JC, Mirin J. Prescribing trends of antidepressants in bipolar depression. J Clin Psychiatry1995; 56:260–264.
Mallinger AG, Frank E, Thase ME, Barwell MM, Diazgranados N, Luckenbaugh DA, Kupfer DJ. Revisiting the effectiveness of standard antidepressants in bipolar disorder: are monoamine oxidase inhibitors superior?Psychopharmacol Bull2009; 42:64–74.
Anderson IM, Nutt DJ, Deakin JF. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 1993 British Association for Psychopharmacology guidelines. British Association for Psychopharmacology. J Psychopharmacol2000; 14:3–20.
Liebowitz MR, Hollander E, Schneier F, et al. Reversible and irreversible monoamine oxidase inhibitors in other psychiatric disorders. Acta Psychiatr Scand Suppl1990; 360:29–34.
Davidson JR, Giller EL, Zisook S, Overall JE. An efficacy study of isocarboxazid and placebo in depression, and its relationship to depressive nosology. Arch Gen Psychiatry1988; 45:120–127.
Razani J, White KL, White J, et al. The safety and efficacy of combined amitriptyline and tranylcypromine antidepressant treatment: a controlled trial. Arch Gen Psychiatry1983; 40:657–661.
Thase ME, Triverdi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology1995; 12:185–219.
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision. Washington, DC, American Psychiatric Association, 2000.
Parker G, Crawford J. Atypical depression: retrospective self-reporting of treatment effectiveness. Acta Psychiatr Scand2009; 120:213–221.
Henkel V, Mergl R, Algaier AK, Kohnen R, Moller HJ, Hergerl U. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res2006; 141:89–101.
Rothschild R, Quitkin HM, Quitkin FM, et al. A double-blind placebo-controlled comparison of phenelzine and imipramine in the treatment of bulimia in atypical depressives. Int J Eat Disord1994; 15:1–9.
Judd LL, Paulus MJ, Schettler PJ, et al. Does incomplete recovery from first life-time major depressive episode herald a chronic course of illness?Am J Psychiatry2000; 157:1501–1504.
Murphy JM, Monson RR, Olivier DC, Sobol AM, Leighton AH. Affective disorders and mortality: a general population study. Arch Gen Psychiatry1987; 44:473–480.
Thase ME, Frank E, Mallinger AG, Hamer T, Kupfer DJ. Treatment of imipramine-resistant recurrent depression, III: efficacy of monoamine oxidase inhibitors. J Clin Psychiatry1992; 53:5–11.
Thase ME, Mallinger AG, McKnight D, Himmelhoch JM. Treatment of imipramine-resistant recurrent depression, IV: a double blind crossover study of tranylcypromine for anergic bipolar depression. Am J Psychiatry1992; 149:195–198.
Quitkin FM, McGrath PJ, Stewart JW, et al. Atypical depression, panic attacks, and response to imipramine and phenelzine: a replication. Arch Gen Psychiatry1990; 47:935–941.
Himmelhoch JM, Thase ME, Mallinger AG, Houck P. Tranylcypromine versus imipramine in anergic bipolar depression. Am J Psychiatry1991; 148:910–916.
Bodkin JA, Amsterdam JD. Transdermal selegiline in major depression: a double-blind, placebo-controlled, parallel-group study in outpatients. Am J Psychiatry2002; 159:1869–1875.
Amsterdam JD. A double-blind, placebo-controlled trial of the safety and efficacy of selegiline transdermal system without dietary restrictions in patients with major depressive disorder. J Clin Psychiatry2003; 64:208–214.
Amsterdam JD, Bodkin JA. Selegiline transdermal system in the prevention of relapse of major depressive disorder: a 52-week, double-blind, placebo-substitution, parallel-group clinical trial. J Clin Psychopharmacol2006; 26:579–586.
Feiger AD, Rickels K, Rynn MA, et al. Selegiline transdermal system for the treatment of major depressive disorder: an 8-week, double-blind, placebo-controlled, flexible-dose titration trial. J Clin Psychiatry2006; 67:1354–1361.
Monoamine oxidase (MAO) inhibitors were the first drugs for treating depression. Introduced in the 1950s, they were used extensively for the next two decades. Their use declined substantially since then because of their reported side effects, their food and drug interactions, and the introduction of new classes of antidepressants.
This trend may be changing. These drugs can be effective in major depressive disorder, and particularly in major depressive disorder with atypical features and in treatment-resistant depression.
New, selective MAO inhibitors are being developed. Moreover, the selegiline transdermal system (Emsam),1,2 introduced in 2006, offers the potential advantage of eliminating the need for burdensome dietary restrictions and has renewed interest in this group of drugs.
In this article, we discuss the history, pharmacology, safety and tolerability of MAO inhibitors, and we summarize recent MAO inhibitor research. Our goal is to familiarize physicians with this class of drugs, including recent updates regarding their safety profile and liberalized dietary recommendations.
DEPRESSION IS COMMON, DIFFICULT
Depression affects 121 million people worldwide.3 According to a study that compared two surveys of 40,000 people each, the prevalence of major depressive disorder in the United States more than doubled (from 3.3% to 7.0%) from 1992 to 2002.4 Another survey, in 2002 and 2003, revealed the lifetime prevalence of major depressive disorder to be 16.6%.5
Treatments for depression have expanded over the past 20 years, with new classes of drugs such as selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs). However, depression has remained a difficult condition to treat. In the National Institute of Mental Health’s Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study,6 the remission rate in patients treated with the SSRI citalopram (Celexa) for up to 14 weeks was 28% using one measure and 33% using another. Diversifying and understanding existing and emerging therapeutic options is important to the effective treatment of this disease.
THE RISE AND FALL OF MAO INHIBITORS
The first antidepressant introduced was an MAO inhibitor, iproniazid, followed shortly thereafter by a tricyclic antidepressant, imipramine (Tofranil). When iproniazid, originally an antituberculosis agent, was promoted for its antidepressant properties in the 1950s, very little was known about its side effects. It was later removed from the market because of hepatotoxicity, but several other MAO inhibitors had surfaced for the treatment of depression—eg, phenelzine (Nardil), isocarboxazid (Marplan), and tranylcypromine (Parnate).
Currently, MAO inhibitors are typically reserved for third- or fourth-line treatment. As a result, even psychiatrists have little experience with these agents. In a 1999 survey of the Michigan Psychiatric Association,7 12% of practicing psychiatrists said they had never prescribed an MAO inhibitor, another 27% had not prescribed one in the prior 3 years, and only 2% said they prescribed them frequently. A decade earlier, about 25% had said they prescribed them often.8
The prescription rate of MAO inhibitors has remained low during the past 10 years. In a Canadian population-based study9 conducted among older adults in a large health care database from January 1997 to April 2007, the yearly incidence of MAO inhibitor prescriptions decreased from a rate of 3.1 per 100,000 to 1.4 per 100,000. Drug interactions, side effects, preference for other treatments, and dietary restrictions were the reasons most often cited for not prescribing these drugs.7
The side effects of MAO inhibitors were recognized by the mid-1960s, when more than 40 cases of tyramine-induced hypertensive crisis were reported (particularly with tranylcypromine).10,11 Many of the reported events happened after the patient ate tyramine-rich foods such as aged cheese (hence, “the cheese reaction”—more on this below) or drank draft beer.10,11 The US Food and Drug Administration (FDA) consequently established dietary restrictions for patients taking MAO inhibitors, but people found the guidelines cumbersome and often switched to newer drugs that did not require a restrictive diet, such as tricyclics and, much later (in the 1980s), SSRIs.
MAO HAS TWO SUBTYPES
MAO is a flavin-containing enzyme critical for regulating neurotransmitter levels by catabolizing endogenous monoamines (eg, norepinephrine, serotonin, and dopamine) and exogenous amines (eg, dietary tyramine). It is found throughout the body but is more highly concentrated in the liver, kidneys, intestinal wall, and brain.
MAO has two subtypes, isoenzyme A (MAO-A) and isoenzyme B (MAO-B), which vary in their distribution. MAO-A is found primarily in the intestinal tract, liver, and peripheral adrenergic neurons (adrenal glands, arterial vessels, and sympathetic nerves) and preferentially metabolizes serotonin and norepinephrine. MAO-B is found mostly in the brain and liver. However, both isotypes are found in all of the areas mentioned. Since 80% of intestinal MAO is MAO-A, this isoenzyme is primarily responsible for degradation of tyramine, and thus inhibition of MAO-A is associated with the cheese reaction.10,11
TYPES OF MAO INHIBITORS
MAO inhibitors can be classified on the basis of whether they are nonselective or selective for either MAO-A or MAO-B, and whether their effect is reversible.
Nonselective MAO inhibitors are phenelzine, isocarboxazid, and tranylcypromine.
Selective MAO inhibitors. Selegiline is selective for MAO-B. Clorgyline is selective for MAO-A, but it is not available in the United States.
A reversible MAO inhibitor is moclobemide (not available in the United States).
Do selectivity and reversibility matter?
Classic MAO inhibitors such as tranylcypromine and phenelzine are neither reversible (binding to the enzyme for the extent of its lifetime of 14–28 days) nor selective for the subtypes. These drugs were used extensively several decades ago to treat atypical depression, anxiety, and phobias. The only selective MAO inhibitor now available in the United States is selegiline, which inhibits MAO-B at low doses but loses its selectivity at dosages greater than 20 mg/day.
Experimental studies suggest that inhibition of more than 70% of MAO-A activity is necessary for the antidepressant effect of selegiline.12 At oral doses that selectively inhibit MAO-B (5–10 mg/day), selegiline does not seem to have potent antidepressant activity, although it does show success as an adjunctive treatment for Parkinson disease and does not necessitate any dietary restriction. Only at higher oral doses (20–60 mg/day), at which MAO-B selectivity is lost, is the antidepressant effect seen. But the higher doses necessitate dietary restrictions. Therefore, patients who are taking the oral selective MAO inhibitor selegiline have to follow the same dietary restrictions as patients taking the nonselective ones.
Reversible inhibitors of MAO-A have the distinction of being easily displaced by ingested tyramine in the gut and thus do not cause the cheese reaction. However, the only reversible agent available in the world market is moclobemide. It is not available in the United States, and appears to be less effective than older, nonselective MAO inhibitors.13
SELEGILINE TRANSDERMAL SYSTEM
The selegiline transdermal system (Emsam) is the first FDA-approved transdermal patch for treatment of major depression. Patients who are using Emsam at its lowest effective dose of 6 mg/24 hours do not need to follow the dietary restrictions that are needed for all oral MAO inhibitors.
Pharmacokinetics of the selegiline patch
With the transdermal patch, selegiline is extensively absorbed through the skin. Plasma levels are maintained over a 24-hour period, allowing once-daily application. Patches are available that deliver 6, 9, or 12 mg per 24 hours. Steady-state plasma levels are reached after about 5 days.
The bioavailability of selegiline is about 75% with the transdermal delivery system vs 4.4% after oral administration, the lower number being due to first-pass metabolism.1 About 90% of selegiline is bound to plasma proteins and quickly penetrates the central nervous system.
This drug is metabolized by cytochrome P450 isoenzymes, including CYP2C9, CYP2B6, and CYP3A4. Its metabolites are l-methamphetamine and n-desmethylselegiline.
Clinical research showed that dosage adjustments were not necessary in specific populations studied, including patients with various stages of renal or hepatic failure.1 Clearance of selegiline was independent of dose, age, sex, renal function, body weight, or concomitant medications.1
Advantages of the patch system
Since selegiline delivered via the patch is not absorbed through the gut, it has little effect on gut MAO-A and therefore is unlikely to lead to tyramine-induced hypertensive crisis. Studies of the selegiline patch show that inhibition of more than 80% of gut MAO-A is necessary to impair metabolism of tyramine in the gut.1 Therefore, the 6-mg patch will not significantly impair tyramine degradation in the gut. In phase III testing of the selegiline patch, no hypertensive crises were reported among 2,656 outpatients without dietary restrictions. However, it is still recommended that patients on the 9-mg and 12-mg patches follow a tyramine-free diet.1
Although there are no data available to suggest that higher dosages are more effective, it is recommended that the dose be titrated in 3-mg increments at intervals of at least 2 weeks until the maximum recommended dosage of 12 mg/24 hours is reached.2
Disadvantage of the selegiline patch: Cost
The selegiline patch is expensive: $692.99 for 1 month’s supply at a dose of 6 mg/24 hours and $638.99 for 1 month’s supply at a dose of 9 or 12 mg/24 hours (verified with a national pharmacy chain at the time of this writing). Insurance coverage for the patch varies, and documentation may be required from the physician. Oral MAO inhibitors are much less expensive.
SAFETY, TOLERABILITY OF MAO INHIBITORS
Side effects of oral agents
Orthostatic hypotension, dizziness, drowsiness, insomnia, and nausea are the most frequently reported side effects of oral MAO inhibitors.14,15 These side effects can generally be managed symptomatically by slowing the titration, dividing the doses, changing the time it is taken, or, in the case of orthostatic hypotension, increasing fluid intake.14 Phenelzine has the strongest association with sedation.14
Weight gain, edema, muscle pain, myoclonus, paresthesias, sexual dysfunction, and, rarely, hepatotoxicity are late side effects.15–18 Paresthesias, an infrequent side effect, are often treated with pyridoxine supplementation.15
Transient hypertensive episodes within 2 hours after ingestion of MAO inhibitors, which were independent of dietary or drug interactions, have been reported.19 The hypertensive episodes are usually self-limited but in rare cases result in hypertensive crisis.19–21
Serotonin syndrome has been reported with MAO inhibitor monotherapy in rare cases.22 Serotonin syndrome is characterized by mental status changes, restlessness, myoclonus, hyperreflexia, diaphoresis, or evidence of autonomic hyperactivity.23 The syndrome is potentially fatal and is treated symptomatically by removing the offending drugs and giving intravenous rehydration.23
Side effects of the selegiline patch
The most common adverse events with the selegiline patch include application-site reaction (24% vs 12% with placebo), headache (18% vs 17%), insomnia, diarrhea, dry mouth, and dyspepsia.24,25 Dose-related orthostatic hypotension was reported (occurring in 9.8% vs 6.7% with placebo) and was most likely to occur in elderly patients.25 It is suggested that insomnia may be lessened by removing the patch before bedtime. Also, rotating the patch application sites and prompt topical treatment of irritation may lessen local effects.24
Observe a washout period when switching between serotonergic drugs
Most MAO inhibitors irreversibly inhibit MAO for the life of the enzyme, and thus the physiologic effects of phenelzine, isocarboxazid, and tranylcypromine last for up to 2 to 3 weeks.26 Although the elimination half-life of typical MAO inhibitors is short (1.5–4 hours),27,28 their physiologic effects are long-lasting.14
Switching from a MAO inhibitor to another serotonergic agent. Concomitant use of MAO inhibitors and other serotonergic drugs is associated with the risk of serotonin syndrome. After stopping an MAO inhibitor, a 14-day washout period is recommended before starting another serotonergic agent.29 Patients should continue to be monitored closely after the washout period, as cases of serotonin syndrome have been reported later.30 A 14-day washout period is also recommended when switching between MAO inhibitors, although more rapid switches have been made safely.31
Switching from another serotonergic agent to an oral MAO inhibitor. Similarly, a 14-day washout period (or five half-lives) is necessary after stopping most of the serotonergic agents mentioned above before beginning treatment with an oral MAO inhibitor. Fluoxetine (Prozac) has a longer half-life and therefore requires a longer washout period, ie, 5 weeks.
Switching from another serotonergic agent to the selegiline patch. When switching to the selegiline patch from another serotonergic drug, the washout period is 1 week after stopping most drugs or 5 weeks after stopping fluoxetine. One must wait 2 weeks after stopping the selegiline patch before starting therapy with any of the other serotonergic drugs.
Drugs to avoid due to interactions
In view of the risk of severe of drug-drug interactions, particularly the risk of serotonin syndrome, the following serotonin-enhancing compounds are contraindicated in patients taking a MAO inhibitor: SSRIs, SNRIs, tricyclic antidepressants, other MAO inhibitors, mirtazapine, and St. John’s wort. Other pharmaceuticals to be avoided include bupropion, meperidine, tramadol, methadone, propoxyphene, pentazocine, dextromethorphan, and cyclobenzaprine (Table 1). Also, there have been numerous reports of serotonin syndrome with the use of the broad-spectrum, MAO-based antibiotic linezolid (Zyvox), by itself or in conjunction with other serotonergic agents.32–35
Several studies suggested a hazardous combination of nonsubcutaneous sumatriptans (5-HT1B/1D agonists) and MAO-B inhibitors, while subcutaneous sumatriptan migraine-abortive treatment and MAO-B inhibitors appear to be safe.36,37
Also, amphetamines, cough-and-cold preparations, and weight-reducing preparations that contain vasoconstrictors (eg, pseudoephedrine, phenylephrine, phenylpropanolamine, and ephedrine) should be avoided, as the risk of hypertensive crisis increases with these products.
Patients on MAO inhibitors should wear a medical alert bracelet in case they need to undergo emergency surgery and are unable to verbally communicate their drug history. They should be instructed to alert all health care providers about their MAO inhibitor use.14
Beware of worsening depression
Physicians, patients, and family members should be advised to observe for worsening depression or “suicidality” during the course of treatment with MAO inhibitors, as with all antidepressants.
Diet can be more lenient than in the past
The dietary restrictions classically advised for patients taking oral MAO inhibitors were established to prevent hypertensive crises associated with tyramine ingestion. However, some of these restrictions were unsubstantiated,38 and evidence from more recent studies suggests that they are unnecessarily strict39 and may lead to resistance by the physician, the patient, or both to using this potentially beneficial therapy.14 There is also a risk that patients will inadvertently discover that a food that was in the “restricted” list caused them no harm upon ingestion and thus will become cavalier about dietary adherence.39
To prevent dietary noncompliance, physicians should conduct ongoing diet surveys and encourage adherence to evidence-based dietary recommendations.40
The FDA and drug-package inserts for oral MAO inhibitors continue to recommend stringent dietary restrictions, including no aged cheeses or meats, soy sauce, soy beans, soy paste, miso soup, Italian green beans (fava beans), snow peas, broad bean pods, sauerkraut, kimchee, concentrated yeast extracts (Marmite), wine, beer (including alcohol-free beer), and many other foods. However, several studies have measured the tyramine content of food and determined that less than 6 mg per serving is generally safe.39,41 The results of these investigations have led to more lenient dietary guidelines.39
Absolute dietary restrictions include39:
Aged cheeses and meats
Banana peels
Broad bean (fava) pods
Spoiled meats
Marmite
Sauerkraut
Soybean products
Draft beers.
Among the many foods determined to be unnecessarily restricted are avocados; bananas; beef or chicken bouillon; chocolate; fresh and mild cheeses, eg, ricotta, cottage cheese, cream cheese, processed cheese slices; fresh meat, poultry, or fish; meat gravy (fresh); monosodium glutamate; peanuts; properly stored pickled or smoked fish (eg, herring); raspberries; and yeast extracts (except Marmite).39
Dietary restrictions should continue for 2 weeks after stopping an MAO inhibitor.
Dietary restrictions for the selegiline patch
Tyramine-containing foods pose less risk with the selegiline patch than with oral MAO inhibitors, and studies42 show that the 6-mg patch does not necessitate dietary restrictions. The accumulating data suggest that the risk of a tyramine-induced event is extremely low with the patch even in doses above 6 mg. But in the meantime, the recommendations for the 9-mg and 12-mg patches remain the same as for the classic oral MAO inhibitors, and tyramine-containing food should be restricted.
EFFICACY OF MAO INHIBITORS IN CLINICAL PRACTICE
Data from numerous studies suggest MAO inhibitors are effective in managing major depressive disorder, and specifically atypical depression,43–48 treatment-resistant major depressive disorder,49,50 and bipolar depression.51,52 Guidelines from the American Psychiatric Association and the British Association for Psychopharmacology suggest that MAO inhibitors be recommended for treatment of major depressive disorder in patients with atypical features and when other antidepressants have failed.53
MAO inhibitors have also been used in the treatment of Parkinson disease, bulimia, anxiety disorders, anorexia nervosa, and body dysmorphic disorder.54
Major depressive disorder
In controlled trials in outpatients with depression who were treated with therapeutic doses of MAO inhibitors, the response rate was 50% to 70%.55 When tranylcypromine was used in severely depressed inpatients, its efficacy was comparable to that of electroconvulsive therapy, imipramine, and amitriptyline.56 Thase et al,57 in a meta-analysis, found that the MAO inhibitors tranylcypromine, phenelzine, and isocarboxazid were equally effective in treating depression.
Atypical depression
Atypical depression is one of the most common subtypes of major depressive disorder. Diagnostic criteria for major depressive disorder with atypical features include mood reactivity and two of the following: weight gain or hyperphagia, hypersomnolence, leaden paralysis, or an enduring pattern of rejection sensitivity.58 An estimated 30% of outpatients with unipolar depression meet these criteria.59
Multiple randomized controlled trials showed that MAO inhibitors were superior to tricyclic antidepressants in treating atypical depression. One study, involving more than 400 patients, determined that atypical depression responded better to phenelzine than to imipramine.43 Another study evaluating 153 critically depressed patients showed significantly greater response with phenelzine than with imipramine or placebo.49 Furthermore, in another double-blind controlled crossover study, 89 mood-reactive, nonmelancholic, chronically depressed outpatients were found to have a striking response to phenelzine after being unresponsive to imipramine.50 Another report48 indicated that in a double-blind, randomized, placebo-controlled trial among 119 patients with atypical depression treated for 6 weeks, the overall response rates were 78% with phenelzine, 50% with imipramine, and 28% with placebo.
A recent meta-analysis of treatment trials in atypical depression revealed a large mean effect size of 0.45 for the superiority of MAO inhibitors over placebo and a medium mean effect size of 0.27 for the superiority of MAO inhibitors over tricyclic antidepressants.60 Additionally, in a randomized, double-blind placebo-controlled trial, patients with comorbid atypical depression and bulimia showed significant improvement in both bulimic and depressive symptoms when given phenelzine vs imipramine or placebo.61
The current data comparing SSRIs and MAO inhibitors in the treatment of atypical depression are limited. The above-mentioned meta-analysis of three such trials (when moclobemide was used in two out of three trials) revealed no significant difference in efficacy.60 However, the authors themselves warned about the limitations of the studies, including low power to detect differences. Parker and Crawford59 compared self-rating of effectiveness of the various previous treatments in patients with depression with and without atypical features using an online survey. The analysis of the responses of 1,934 patients showed no overall difference in treatment response to both drug and non-drug therapies between respondents with and without atypical features, except with SSRIs. The “atypical” group had a significantly lower mean effectiveness score for SSRIs overall, and a lower mean effectiveness rating for two of six SSRIs examined. The authors speculated that even though there was no differential outcome detected in individuals with atypical depression treated with MAO inhibitors, this negative finding may simply have reflected the low prevalence of sample respondents who received MAO inhibitors (which was 4% in the “atypical depression” group of 338).59
Treatment-resistant depression
The ultimate goal in treating major depressive disorder is to achieve complete remission. If complete remission is not achieved, the risk of relapse is high,62,63 as is the risk of more severe future depressive episodes63 and death from any cause.64 Therefore, the ability of clinicians to make appropriate and evidence-based changes in treatment strategy is of high importance.
The use of MAO inhibitors as a third-line or fourth-line choice for treatment-resistant depression is supported by a number of studies.49,50,65–67 MAO inhibitors appear to be especially effective in the subgroup of patients who have treatment-resistant depression with atypical or anergic bipolar features.
Bipolar depression
Anergic bipolar depression is defined as a condition associated with fatigue, psychomotor retardation, and at least one reversed neurovegetative symptom in a patient with bipolar disorder meeting the criteria for a major depressive episode. According to several trials,51,52,68 MAO inhibitors may be more effective than a tricyclic antidepressant in the treatment of anergic bipolar depression. However, more studies are required to determine the role of antidepressants in general and MAO inhibitors in particular in the management of bipolar depression.
Efficacy of the selegiline patch
The efficacy of the selegiline patch in the treatment of depression was examined in four double-blind placebo-controlled studies.69–72 There were three short-term studies (a 6-week study of 177 patients,69 an 8-week study of 265 patients,72 and an 8-week study of 289 patients70) and a fixed-dose 1-year relapse prevention study of 322 patients.71 The inclusion criterion for the short-term studies was diagnosis of a first or a recurrent episode of major depressive disorder in patients with a Hamilton Depression Rating Scale (HDRS) score higher than 20. The HDRS score was used to assess improvement in depressive symptoms. In all studies, patients on active patch had significant improvement in depressive symptoms on the HDRS compared with placebo. In the relapse prevention study,71 patients with major depressive disorder that responded to transdermal selegiline 6 mg within the first 10 weeks were stratified either to continue receiving the selegiline 6-mg patch or to receive placebo. Those continually receiving selegiline experienced a significantly longer time to relapse. At 12 months, the relapse rate was 16.8% with the selegiline patch vs 30.7% with placebo. The patch was reported to be well tolerated, with the most common side effect being application site reaction. The adherence to the treatment was high—84.2% in the active-patch group and 89.6% in the placebo group.71
DO MAO INHIBITORS HAVE A PLACE IN PRIMARY CARE?
MAO inhibitors have secured their place in the history of psychiatry as the first antidepressants. Overall, MAO inhibitors remain underused. However, with the introduction of new and selective MAO inhibitors including the selegiline patch, and with data suggesting efficacy in the management of certain subtypes of depression, we expect that interest in this class of drugs will grow among psychiatrists. Based on the current guidelines for MAO inhibitors to be used as a third- or fourth-line treatment, as well as on research data, it is premature to recommend their more extensive use in a primary care setting. Whether this will change in the future depends on both the research advances and new, safer formulations of MAO inhibitors.
Monoamine oxidase (MAO) inhibitors were the first drugs for treating depression. Introduced in the 1950s, they were used extensively for the next two decades. Their use declined substantially since then because of their reported side effects, their food and drug interactions, and the introduction of new classes of antidepressants.
This trend may be changing. These drugs can be effective in major depressive disorder, and particularly in major depressive disorder with atypical features and in treatment-resistant depression.
New, selective MAO inhibitors are being developed. Moreover, the selegiline transdermal system (Emsam),1,2 introduced in 2006, offers the potential advantage of eliminating the need for burdensome dietary restrictions and has renewed interest in this group of drugs.
In this article, we discuss the history, pharmacology, safety and tolerability of MAO inhibitors, and we summarize recent MAO inhibitor research. Our goal is to familiarize physicians with this class of drugs, including recent updates regarding their safety profile and liberalized dietary recommendations.
DEPRESSION IS COMMON, DIFFICULT
Depression affects 121 million people worldwide.3 According to a study that compared two surveys of 40,000 people each, the prevalence of major depressive disorder in the United States more than doubled (from 3.3% to 7.0%) from 1992 to 2002.4 Another survey, in 2002 and 2003, revealed the lifetime prevalence of major depressive disorder to be 16.6%.5
Treatments for depression have expanded over the past 20 years, with new classes of drugs such as selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs). However, depression has remained a difficult condition to treat. In the National Institute of Mental Health’s Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study,6 the remission rate in patients treated with the SSRI citalopram (Celexa) for up to 14 weeks was 28% using one measure and 33% using another. Diversifying and understanding existing and emerging therapeutic options is important to the effective treatment of this disease.
THE RISE AND FALL OF MAO INHIBITORS
The first antidepressant introduced was an MAO inhibitor, iproniazid, followed shortly thereafter by a tricyclic antidepressant, imipramine (Tofranil). When iproniazid, originally an antituberculosis agent, was promoted for its antidepressant properties in the 1950s, very little was known about its side effects. It was later removed from the market because of hepatotoxicity, but several other MAO inhibitors had surfaced for the treatment of depression—eg, phenelzine (Nardil), isocarboxazid (Marplan), and tranylcypromine (Parnate).
Currently, MAO inhibitors are typically reserved for third- or fourth-line treatment. As a result, even psychiatrists have little experience with these agents. In a 1999 survey of the Michigan Psychiatric Association,7 12% of practicing psychiatrists said they had never prescribed an MAO inhibitor, another 27% had not prescribed one in the prior 3 years, and only 2% said they prescribed them frequently. A decade earlier, about 25% had said they prescribed them often.8
The prescription rate of MAO inhibitors has remained low during the past 10 years. In a Canadian population-based study9 conducted among older adults in a large health care database from January 1997 to April 2007, the yearly incidence of MAO inhibitor prescriptions decreased from a rate of 3.1 per 100,000 to 1.4 per 100,000. Drug interactions, side effects, preference for other treatments, and dietary restrictions were the reasons most often cited for not prescribing these drugs.7
The side effects of MAO inhibitors were recognized by the mid-1960s, when more than 40 cases of tyramine-induced hypertensive crisis were reported (particularly with tranylcypromine).10,11 Many of the reported events happened after the patient ate tyramine-rich foods such as aged cheese (hence, “the cheese reaction”—more on this below) or drank draft beer.10,11 The US Food and Drug Administration (FDA) consequently established dietary restrictions for patients taking MAO inhibitors, but people found the guidelines cumbersome and often switched to newer drugs that did not require a restrictive diet, such as tricyclics and, much later (in the 1980s), SSRIs.
MAO HAS TWO SUBTYPES
MAO is a flavin-containing enzyme critical for regulating neurotransmitter levels by catabolizing endogenous monoamines (eg, norepinephrine, serotonin, and dopamine) and exogenous amines (eg, dietary tyramine). It is found throughout the body but is more highly concentrated in the liver, kidneys, intestinal wall, and brain.
MAO has two subtypes, isoenzyme A (MAO-A) and isoenzyme B (MAO-B), which vary in their distribution. MAO-A is found primarily in the intestinal tract, liver, and peripheral adrenergic neurons (adrenal glands, arterial vessels, and sympathetic nerves) and preferentially metabolizes serotonin and norepinephrine. MAO-B is found mostly in the brain and liver. However, both isotypes are found in all of the areas mentioned. Since 80% of intestinal MAO is MAO-A, this isoenzyme is primarily responsible for degradation of tyramine, and thus inhibition of MAO-A is associated with the cheese reaction.10,11
TYPES OF MAO INHIBITORS
MAO inhibitors can be classified on the basis of whether they are nonselective or selective for either MAO-A or MAO-B, and whether their effect is reversible.
Nonselective MAO inhibitors are phenelzine, isocarboxazid, and tranylcypromine.
Selective MAO inhibitors. Selegiline is selective for MAO-B. Clorgyline is selective for MAO-A, but it is not available in the United States.
A reversible MAO inhibitor is moclobemide (not available in the United States).
Do selectivity and reversibility matter?
Classic MAO inhibitors such as tranylcypromine and phenelzine are neither reversible (binding to the enzyme for the extent of its lifetime of 14–28 days) nor selective for the subtypes. These drugs were used extensively several decades ago to treat atypical depression, anxiety, and phobias. The only selective MAO inhibitor now available in the United States is selegiline, which inhibits MAO-B at low doses but loses its selectivity at dosages greater than 20 mg/day.
Experimental studies suggest that inhibition of more than 70% of MAO-A activity is necessary for the antidepressant effect of selegiline.12 At oral doses that selectively inhibit MAO-B (5–10 mg/day), selegiline does not seem to have potent antidepressant activity, although it does show success as an adjunctive treatment for Parkinson disease and does not necessitate any dietary restriction. Only at higher oral doses (20–60 mg/day), at which MAO-B selectivity is lost, is the antidepressant effect seen. But the higher doses necessitate dietary restrictions. Therefore, patients who are taking the oral selective MAO inhibitor selegiline have to follow the same dietary restrictions as patients taking the nonselective ones.
Reversible inhibitors of MAO-A have the distinction of being easily displaced by ingested tyramine in the gut and thus do not cause the cheese reaction. However, the only reversible agent available in the world market is moclobemide. It is not available in the United States, and appears to be less effective than older, nonselective MAO inhibitors.13
SELEGILINE TRANSDERMAL SYSTEM
The selegiline transdermal system (Emsam) is the first FDA-approved transdermal patch for treatment of major depression. Patients who are using Emsam at its lowest effective dose of 6 mg/24 hours do not need to follow the dietary restrictions that are needed for all oral MAO inhibitors.
Pharmacokinetics of the selegiline patch
With the transdermal patch, selegiline is extensively absorbed through the skin. Plasma levels are maintained over a 24-hour period, allowing once-daily application. Patches are available that deliver 6, 9, or 12 mg per 24 hours. Steady-state plasma levels are reached after about 5 days.
The bioavailability of selegiline is about 75% with the transdermal delivery system vs 4.4% after oral administration, the lower number being due to first-pass metabolism.1 About 90% of selegiline is bound to plasma proteins and quickly penetrates the central nervous system.
This drug is metabolized by cytochrome P450 isoenzymes, including CYP2C9, CYP2B6, and CYP3A4. Its metabolites are l-methamphetamine and n-desmethylselegiline.
Clinical research showed that dosage adjustments were not necessary in specific populations studied, including patients with various stages of renal or hepatic failure.1 Clearance of selegiline was independent of dose, age, sex, renal function, body weight, or concomitant medications.1
Advantages of the patch system
Since selegiline delivered via the patch is not absorbed through the gut, it has little effect on gut MAO-A and therefore is unlikely to lead to tyramine-induced hypertensive crisis. Studies of the selegiline patch show that inhibition of more than 80% of gut MAO-A is necessary to impair metabolism of tyramine in the gut.1 Therefore, the 6-mg patch will not significantly impair tyramine degradation in the gut. In phase III testing of the selegiline patch, no hypertensive crises were reported among 2,656 outpatients without dietary restrictions. However, it is still recommended that patients on the 9-mg and 12-mg patches follow a tyramine-free diet.1
Although there are no data available to suggest that higher dosages are more effective, it is recommended that the dose be titrated in 3-mg increments at intervals of at least 2 weeks until the maximum recommended dosage of 12 mg/24 hours is reached.2
Disadvantage of the selegiline patch: Cost
The selegiline patch is expensive: $692.99 for 1 month’s supply at a dose of 6 mg/24 hours and $638.99 for 1 month’s supply at a dose of 9 or 12 mg/24 hours (verified with a national pharmacy chain at the time of this writing). Insurance coverage for the patch varies, and documentation may be required from the physician. Oral MAO inhibitors are much less expensive.
SAFETY, TOLERABILITY OF MAO INHIBITORS
Side effects of oral agents
Orthostatic hypotension, dizziness, drowsiness, insomnia, and nausea are the most frequently reported side effects of oral MAO inhibitors.14,15 These side effects can generally be managed symptomatically by slowing the titration, dividing the doses, changing the time it is taken, or, in the case of orthostatic hypotension, increasing fluid intake.14 Phenelzine has the strongest association with sedation.14
Weight gain, edema, muscle pain, myoclonus, paresthesias, sexual dysfunction, and, rarely, hepatotoxicity are late side effects.15–18 Paresthesias, an infrequent side effect, are often treated with pyridoxine supplementation.15
Transient hypertensive episodes within 2 hours after ingestion of MAO inhibitors, which were independent of dietary or drug interactions, have been reported.19 The hypertensive episodes are usually self-limited but in rare cases result in hypertensive crisis.19–21
Serotonin syndrome has been reported with MAO inhibitor monotherapy in rare cases.22 Serotonin syndrome is characterized by mental status changes, restlessness, myoclonus, hyperreflexia, diaphoresis, or evidence of autonomic hyperactivity.23 The syndrome is potentially fatal and is treated symptomatically by removing the offending drugs and giving intravenous rehydration.23
Side effects of the selegiline patch
The most common adverse events with the selegiline patch include application-site reaction (24% vs 12% with placebo), headache (18% vs 17%), insomnia, diarrhea, dry mouth, and dyspepsia.24,25 Dose-related orthostatic hypotension was reported (occurring in 9.8% vs 6.7% with placebo) and was most likely to occur in elderly patients.25 It is suggested that insomnia may be lessened by removing the patch before bedtime. Also, rotating the patch application sites and prompt topical treatment of irritation may lessen local effects.24
Observe a washout period when switching between serotonergic drugs
Most MAO inhibitors irreversibly inhibit MAO for the life of the enzyme, and thus the physiologic effects of phenelzine, isocarboxazid, and tranylcypromine last for up to 2 to 3 weeks.26 Although the elimination half-life of typical MAO inhibitors is short (1.5–4 hours),27,28 their physiologic effects are long-lasting.14
Switching from a MAO inhibitor to another serotonergic agent. Concomitant use of MAO inhibitors and other serotonergic drugs is associated with the risk of serotonin syndrome. After stopping an MAO inhibitor, a 14-day washout period is recommended before starting another serotonergic agent.29 Patients should continue to be monitored closely after the washout period, as cases of serotonin syndrome have been reported later.30 A 14-day washout period is also recommended when switching between MAO inhibitors, although more rapid switches have been made safely.31
Switching from another serotonergic agent to an oral MAO inhibitor. Similarly, a 14-day washout period (or five half-lives) is necessary after stopping most of the serotonergic agents mentioned above before beginning treatment with an oral MAO inhibitor. Fluoxetine (Prozac) has a longer half-life and therefore requires a longer washout period, ie, 5 weeks.
Switching from another serotonergic agent to the selegiline patch. When switching to the selegiline patch from another serotonergic drug, the washout period is 1 week after stopping most drugs or 5 weeks after stopping fluoxetine. One must wait 2 weeks after stopping the selegiline patch before starting therapy with any of the other serotonergic drugs.
Drugs to avoid due to interactions
In view of the risk of severe of drug-drug interactions, particularly the risk of serotonin syndrome, the following serotonin-enhancing compounds are contraindicated in patients taking a MAO inhibitor: SSRIs, SNRIs, tricyclic antidepressants, other MAO inhibitors, mirtazapine, and St. John’s wort. Other pharmaceuticals to be avoided include bupropion, meperidine, tramadol, methadone, propoxyphene, pentazocine, dextromethorphan, and cyclobenzaprine (Table 1). Also, there have been numerous reports of serotonin syndrome with the use of the broad-spectrum, MAO-based antibiotic linezolid (Zyvox), by itself or in conjunction with other serotonergic agents.32–35
Several studies suggested a hazardous combination of nonsubcutaneous sumatriptans (5-HT1B/1D agonists) and MAO-B inhibitors, while subcutaneous sumatriptan migraine-abortive treatment and MAO-B inhibitors appear to be safe.36,37
Also, amphetamines, cough-and-cold preparations, and weight-reducing preparations that contain vasoconstrictors (eg, pseudoephedrine, phenylephrine, phenylpropanolamine, and ephedrine) should be avoided, as the risk of hypertensive crisis increases with these products.
Patients on MAO inhibitors should wear a medical alert bracelet in case they need to undergo emergency surgery and are unable to verbally communicate their drug history. They should be instructed to alert all health care providers about their MAO inhibitor use.14
Beware of worsening depression
Physicians, patients, and family members should be advised to observe for worsening depression or “suicidality” during the course of treatment with MAO inhibitors, as with all antidepressants.
Diet can be more lenient than in the past
The dietary restrictions classically advised for patients taking oral MAO inhibitors were established to prevent hypertensive crises associated with tyramine ingestion. However, some of these restrictions were unsubstantiated,38 and evidence from more recent studies suggests that they are unnecessarily strict39 and may lead to resistance by the physician, the patient, or both to using this potentially beneficial therapy.14 There is also a risk that patients will inadvertently discover that a food that was in the “restricted” list caused them no harm upon ingestion and thus will become cavalier about dietary adherence.39
To prevent dietary noncompliance, physicians should conduct ongoing diet surveys and encourage adherence to evidence-based dietary recommendations.40
The FDA and drug-package inserts for oral MAO inhibitors continue to recommend stringent dietary restrictions, including no aged cheeses or meats, soy sauce, soy beans, soy paste, miso soup, Italian green beans (fava beans), snow peas, broad bean pods, sauerkraut, kimchee, concentrated yeast extracts (Marmite), wine, beer (including alcohol-free beer), and many other foods. However, several studies have measured the tyramine content of food and determined that less than 6 mg per serving is generally safe.39,41 The results of these investigations have led to more lenient dietary guidelines.39
Absolute dietary restrictions include39:
Aged cheeses and meats
Banana peels
Broad bean (fava) pods
Spoiled meats
Marmite
Sauerkraut
Soybean products
Draft beers.
Among the many foods determined to be unnecessarily restricted are avocados; bananas; beef or chicken bouillon; chocolate; fresh and mild cheeses, eg, ricotta, cottage cheese, cream cheese, processed cheese slices; fresh meat, poultry, or fish; meat gravy (fresh); monosodium glutamate; peanuts; properly stored pickled or smoked fish (eg, herring); raspberries; and yeast extracts (except Marmite).39
Dietary restrictions should continue for 2 weeks after stopping an MAO inhibitor.
Dietary restrictions for the selegiline patch
Tyramine-containing foods pose less risk with the selegiline patch than with oral MAO inhibitors, and studies42 show that the 6-mg patch does not necessitate dietary restrictions. The accumulating data suggest that the risk of a tyramine-induced event is extremely low with the patch even in doses above 6 mg. But in the meantime, the recommendations for the 9-mg and 12-mg patches remain the same as for the classic oral MAO inhibitors, and tyramine-containing food should be restricted.
EFFICACY OF MAO INHIBITORS IN CLINICAL PRACTICE
Data from numerous studies suggest MAO inhibitors are effective in managing major depressive disorder, and specifically atypical depression,43–48 treatment-resistant major depressive disorder,49,50 and bipolar depression.51,52 Guidelines from the American Psychiatric Association and the British Association for Psychopharmacology suggest that MAO inhibitors be recommended for treatment of major depressive disorder in patients with atypical features and when other antidepressants have failed.53
MAO inhibitors have also been used in the treatment of Parkinson disease, bulimia, anxiety disorders, anorexia nervosa, and body dysmorphic disorder.54
Major depressive disorder
In controlled trials in outpatients with depression who were treated with therapeutic doses of MAO inhibitors, the response rate was 50% to 70%.55 When tranylcypromine was used in severely depressed inpatients, its efficacy was comparable to that of electroconvulsive therapy, imipramine, and amitriptyline.56 Thase et al,57 in a meta-analysis, found that the MAO inhibitors tranylcypromine, phenelzine, and isocarboxazid were equally effective in treating depression.
Atypical depression
Atypical depression is one of the most common subtypes of major depressive disorder. Diagnostic criteria for major depressive disorder with atypical features include mood reactivity and two of the following: weight gain or hyperphagia, hypersomnolence, leaden paralysis, or an enduring pattern of rejection sensitivity.58 An estimated 30% of outpatients with unipolar depression meet these criteria.59
Multiple randomized controlled trials showed that MAO inhibitors were superior to tricyclic antidepressants in treating atypical depression. One study, involving more than 400 patients, determined that atypical depression responded better to phenelzine than to imipramine.43 Another study evaluating 153 critically depressed patients showed significantly greater response with phenelzine than with imipramine or placebo.49 Furthermore, in another double-blind controlled crossover study, 89 mood-reactive, nonmelancholic, chronically depressed outpatients were found to have a striking response to phenelzine after being unresponsive to imipramine.50 Another report48 indicated that in a double-blind, randomized, placebo-controlled trial among 119 patients with atypical depression treated for 6 weeks, the overall response rates were 78% with phenelzine, 50% with imipramine, and 28% with placebo.
A recent meta-analysis of treatment trials in atypical depression revealed a large mean effect size of 0.45 for the superiority of MAO inhibitors over placebo and a medium mean effect size of 0.27 for the superiority of MAO inhibitors over tricyclic antidepressants.60 Additionally, in a randomized, double-blind placebo-controlled trial, patients with comorbid atypical depression and bulimia showed significant improvement in both bulimic and depressive symptoms when given phenelzine vs imipramine or placebo.61
The current data comparing SSRIs and MAO inhibitors in the treatment of atypical depression are limited. The above-mentioned meta-analysis of three such trials (when moclobemide was used in two out of three trials) revealed no significant difference in efficacy.60 However, the authors themselves warned about the limitations of the studies, including low power to detect differences. Parker and Crawford59 compared self-rating of effectiveness of the various previous treatments in patients with depression with and without atypical features using an online survey. The analysis of the responses of 1,934 patients showed no overall difference in treatment response to both drug and non-drug therapies between respondents with and without atypical features, except with SSRIs. The “atypical” group had a significantly lower mean effectiveness score for SSRIs overall, and a lower mean effectiveness rating for two of six SSRIs examined. The authors speculated that even though there was no differential outcome detected in individuals with atypical depression treated with MAO inhibitors, this negative finding may simply have reflected the low prevalence of sample respondents who received MAO inhibitors (which was 4% in the “atypical depression” group of 338).59
Treatment-resistant depression
The ultimate goal in treating major depressive disorder is to achieve complete remission. If complete remission is not achieved, the risk of relapse is high,62,63 as is the risk of more severe future depressive episodes63 and death from any cause.64 Therefore, the ability of clinicians to make appropriate and evidence-based changes in treatment strategy is of high importance.
The use of MAO inhibitors as a third-line or fourth-line choice for treatment-resistant depression is supported by a number of studies.49,50,65–67 MAO inhibitors appear to be especially effective in the subgroup of patients who have treatment-resistant depression with atypical or anergic bipolar features.
Bipolar depression
Anergic bipolar depression is defined as a condition associated with fatigue, psychomotor retardation, and at least one reversed neurovegetative symptom in a patient with bipolar disorder meeting the criteria for a major depressive episode. According to several trials,51,52,68 MAO inhibitors may be more effective than a tricyclic antidepressant in the treatment of anergic bipolar depression. However, more studies are required to determine the role of antidepressants in general and MAO inhibitors in particular in the management of bipolar depression.
Efficacy of the selegiline patch
The efficacy of the selegiline patch in the treatment of depression was examined in four double-blind placebo-controlled studies.69–72 There were three short-term studies (a 6-week study of 177 patients,69 an 8-week study of 265 patients,72 and an 8-week study of 289 patients70) and a fixed-dose 1-year relapse prevention study of 322 patients.71 The inclusion criterion for the short-term studies was diagnosis of a first or a recurrent episode of major depressive disorder in patients with a Hamilton Depression Rating Scale (HDRS) score higher than 20. The HDRS score was used to assess improvement in depressive symptoms. In all studies, patients on active patch had significant improvement in depressive symptoms on the HDRS compared with placebo. In the relapse prevention study,71 patients with major depressive disorder that responded to transdermal selegiline 6 mg within the first 10 weeks were stratified either to continue receiving the selegiline 6-mg patch or to receive placebo. Those continually receiving selegiline experienced a significantly longer time to relapse. At 12 months, the relapse rate was 16.8% with the selegiline patch vs 30.7% with placebo. The patch was reported to be well tolerated, with the most common side effect being application site reaction. The adherence to the treatment was high—84.2% in the active-patch group and 89.6% in the placebo group.71
DO MAO INHIBITORS HAVE A PLACE IN PRIMARY CARE?
MAO inhibitors have secured their place in the history of psychiatry as the first antidepressants. Overall, MAO inhibitors remain underused. However, with the introduction of new and selective MAO inhibitors including the selegiline patch, and with data suggesting efficacy in the management of certain subtypes of depression, we expect that interest in this class of drugs will grow among psychiatrists. Based on the current guidelines for MAO inhibitors to be used as a third- or fourth-line treatment, as well as on research data, it is premature to recommend their more extensive use in a primary care setting. Whether this will change in the future depends on both the research advances and new, safer formulations of MAO inhibitors.
Compton WM, Conway KP, Stinson FS, Grant BF. Changes in the prevalence of major depression and comorbid substance use disorders in the United States between 1991–1992 and 2001–2002. Am J Psychiatry2006; 163:2141–2147.
Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry2005; 62:593–602.
Trivedi MH, Rush AJ, Wisniewski SR, et al; STAR*D Study Team. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry2006; 163:28–40.
Balon R, Mufti R, Arfken CL. A survey of prescribing practices for monoamine oxidase inhibitors. Psychiatr Serv1999; 50:945–947.
Clary C, Mandos LA, Schweizer E. Results of a brief survey on the prescribing practices for monoamine oxidase inhibitor antidepressants. J Clin Psychiatry1990; 51:226–231.
Shulman KI, Fischer HD, Herrmann N, Huo CY, Anderson GM, Rochon PA. Current prescription patterns and safety profile of irreversible monoamine oxidase inhibitors: a population-based cohort study of older adults. J Clin Psychiatry2009; 70:1681–1686.
Horwitz D, Lovenberg W, Engelman K, Sjoerdsma A. Monoamine oxidase inhibitors, tyramine, and cheese. JAMA1964; 188:1108–1110.
Asatoor AM, Levi AJ, Milne MD. Tranylcypromine and cheese. Lancet1963; 2:733–734.
Gordon MN, Muller CD, Sherman KA, Morgan DG, Azzaro AJ, Wecker L. Oral versus transdermal selegiline: antidepressant-like activity in rats. Pharmacol Biochem Behav1999; 63:501–506.
Lotufu-Neto F, Trivedi M, Thase ME. Meta-analysis of the reversible inhibitors of monoamine oxidase type A moclobemide and brofaromine for the treatment of depression. Neuropsychopharmacology1999; 20:226–247.
Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract2004; 10:239–248.
Evans DL, Davidson J, Raft D. Early and late side effects of phenelzine. J Clin Psychopharmacol1982; 2:208–210.
Fava M. Weight gain and antidepressants. J Clin Psychiatry2000; 61(suppl 11):37–41.
Rabkin J, Quitkin F, Harrison W, Tricamo E, McGrath P. Adverse reactions to monoamine oxidase inhibitors. Part I. A comparative study. J Clin Psychopharmacol1984; 4:270–278.
Gomez-Gil E, Salmeron JM, Mas A. Phenelzine-induced fulminant hepatic failure. Ann Intern Med1996; 124:692–693.
Lavin MR, Mendelwitz A, Kronig MH. Spontaneous hypertensive reactions with monoamine oxidase inhibitors. Biol Psychiatry1993; 34:146–151.
Fisher P. Serotonin syndrome in the elderly after antidepressive monotherapy. J Clin Psychopharmacol1995; 15:440–442.
Sternback H. The serotonin syndrome. Am J Psychiatry1991; 148:705–713.
Thase M. Novel transdermal delivery formulation of the monoamine oxidase inhibitor selegiline nearing release for treatment of depression. J Clin Psychiatry2006; 67:671–672.
Lee KC, Chen JJ. Transdermal selegiline for the treatment of major depressive disorder. Neuropsychiatr Dis Treat2007; 3:527–537.
Cooper AJ. Tyramine and irreversible monoamine oxidase inhibitors in clinical practice. Br J Psychiatry Suppl1989; Oct(6):38–45.
Mallinger AG, Smith E. Pharmacokinetics of monoamine oxidase inhibitors. Psychopharmacol Bull1991; 27:493–502.
Fulton B, Benfield P. Moclobemide. An update of its pharmacological properties and therapeutic use. Drugs1996; 52:450–474.
Marangell LB. Switching antidepressants for treatment-resistant major depression. J Clin Psychiatry2001; 62(suppl 18):12–17.
Gitlin MJ. Venlafaxine, monoamine oxidase inhibitors, and the serotonin syndrome. J Clin Psychopharmacol1997; 17:66–67.
Szuba MP, Hornig-Rohan M, Amsterdam JD. Rapid conversion from one monoamine oxidase inhibitor to another. J Clin Psychiatry1997; 58:307–310.
Lorenz RA, Vandenberg AM, Canepa EA. Serotonergic antidepressants and linezolid: a retrospective chart review and presentation of cases. Int J Psychiatry Med2008; 38:81–90.
Miller DG, Lovell EO. Antibiotic-induced serotonin syndrome. J Emerg Med2008, May1(Epub ahead of print).
Das PK, Wakentin DI, Hewko R, Forrest DL. Serotonin syndrome after concomitant treatment with linezolid and meperidine. Clin Infect Dis2008; 46:264–265.
Packer S, Berman SA. Serotonin syndrome precipitated by the monoamine oxidase inhibitor linezolid. Am J Psychiatry2007; 164:346–347.
Diamond S. The use of sumatriptan in patients on monoamine oxidase inhibitors. Neurology1995; 45:1039–1040.
Fox AW. Subcutaneous sumatriptan pharmacokinetics: delimiting the monoamine oxidase inhibitor effect. Headache2010; 50:249–255.
Gardner DM, Shulman KI, Walker SE, Tailor SA. The making of a user friendly MAOI diet. J Clin Psychiatry1996; 57:99–104.
Sweet RA, Brown EJ, Heimberg RG, et al. Monoamine oxidase inhibitor dietary restrictions: what are we asking patients to give up?J Clin Psychiatry1995; 56:196–201.
Shulman KI, Walker SE. Refining the MAOI diet: tyramine content of pizzas and soy products. J Clin Psychiatry1999; 60:191–193.
Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol2006; 46:933–944.
Quitkin FM, Stewart JW, McGrath PJ, et al. Columbia atypical depression. A subgroup of depressives with better response to MAOI than to tricyclic antidepressants or placebo. Br J Psychiatry Suppl1993; Sep(21):30–34.
Davidson J, Pelton S. Forms of atypical depression and their response to antidepressant drugs. Psychiatry Res1986; 17:87–95.
Liebowitz MR, Quitkin FM, Stewart JW, et al. Antidepressant specificity in atypical depression. Arch Gen Psychiatry1988; 45:129–137.
Rapaport MH, Thase ME. Translating the evidence on atypical depression into clinical practice. J Clin Psychiatry2007; 68:e11.
Liebowitz MR. Depression with anxiety and atypical depression. J Clin Psychiatry1993; 54(suppl):10–14.
Stewart JW, McGrath PJ, Quitkin FM, et al. Chronic depression: response to placebo, imipramine, and phenelzine. J Clin Psychopharmacol1993; 13:391–396.
McGrath PJ, Stewart JW, Nunes EV, et al. A double-blind crossover trial of imipramine and phenelzine for outpatients with treatment-refractory depression. Am J Psychiatry1993; 150:118–123.
Zarate CA, Tohen M, Baraibar G, Kando JC, Mirin J. Prescribing trends of antidepressants in bipolar depression. J Clin Psychiatry1995; 56:260–264.
Mallinger AG, Frank E, Thase ME, Barwell MM, Diazgranados N, Luckenbaugh DA, Kupfer DJ. Revisiting the effectiveness of standard antidepressants in bipolar disorder: are monoamine oxidase inhibitors superior?Psychopharmacol Bull2009; 42:64–74.
Anderson IM, Nutt DJ, Deakin JF. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 1993 British Association for Psychopharmacology guidelines. British Association for Psychopharmacology. J Psychopharmacol2000; 14:3–20.
Liebowitz MR, Hollander E, Schneier F, et al. Reversible and irreversible monoamine oxidase inhibitors in other psychiatric disorders. Acta Psychiatr Scand Suppl1990; 360:29–34.
Davidson JR, Giller EL, Zisook S, Overall JE. An efficacy study of isocarboxazid and placebo in depression, and its relationship to depressive nosology. Arch Gen Psychiatry1988; 45:120–127.
Razani J, White KL, White J, et al. The safety and efficacy of combined amitriptyline and tranylcypromine antidepressant treatment: a controlled trial. Arch Gen Psychiatry1983; 40:657–661.
Thase ME, Triverdi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology1995; 12:185–219.
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision. Washington, DC, American Psychiatric Association, 2000.
Parker G, Crawford J. Atypical depression: retrospective self-reporting of treatment effectiveness. Acta Psychiatr Scand2009; 120:213–221.
Henkel V, Mergl R, Algaier AK, Kohnen R, Moller HJ, Hergerl U. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res2006; 141:89–101.
Rothschild R, Quitkin HM, Quitkin FM, et al. A double-blind placebo-controlled comparison of phenelzine and imipramine in the treatment of bulimia in atypical depressives. Int J Eat Disord1994; 15:1–9.
Judd LL, Paulus MJ, Schettler PJ, et al. Does incomplete recovery from first life-time major depressive episode herald a chronic course of illness?Am J Psychiatry2000; 157:1501–1504.
Murphy JM, Monson RR, Olivier DC, Sobol AM, Leighton AH. Affective disorders and mortality: a general population study. Arch Gen Psychiatry1987; 44:473–480.
Thase ME, Frank E, Mallinger AG, Hamer T, Kupfer DJ. Treatment of imipramine-resistant recurrent depression, III: efficacy of monoamine oxidase inhibitors. J Clin Psychiatry1992; 53:5–11.
Thase ME, Mallinger AG, McKnight D, Himmelhoch JM. Treatment of imipramine-resistant recurrent depression, IV: a double blind crossover study of tranylcypromine for anergic bipolar depression. Am J Psychiatry1992; 149:195–198.
Quitkin FM, McGrath PJ, Stewart JW, et al. Atypical depression, panic attacks, and response to imipramine and phenelzine: a replication. Arch Gen Psychiatry1990; 47:935–941.
Himmelhoch JM, Thase ME, Mallinger AG, Houck P. Tranylcypromine versus imipramine in anergic bipolar depression. Am J Psychiatry1991; 148:910–916.
Bodkin JA, Amsterdam JD. Transdermal selegiline in major depression: a double-blind, placebo-controlled, parallel-group study in outpatients. Am J Psychiatry2002; 159:1869–1875.
Amsterdam JD. A double-blind, placebo-controlled trial of the safety and efficacy of selegiline transdermal system without dietary restrictions in patients with major depressive disorder. J Clin Psychiatry2003; 64:208–214.
Amsterdam JD, Bodkin JA. Selegiline transdermal system in the prevention of relapse of major depressive disorder: a 52-week, double-blind, placebo-substitution, parallel-group clinical trial. J Clin Psychopharmacol2006; 26:579–586.
Feiger AD, Rickels K, Rynn MA, et al. Selegiline transdermal system for the treatment of major depressive disorder: an 8-week, double-blind, placebo-controlled, flexible-dose titration trial. J Clin Psychiatry2006; 67:1354–1361.
Compton WM, Conway KP, Stinson FS, Grant BF. Changes in the prevalence of major depression and comorbid substance use disorders in the United States between 1991–1992 and 2001–2002. Am J Psychiatry2006; 163:2141–2147.
Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry2005; 62:593–602.
Trivedi MH, Rush AJ, Wisniewski SR, et al; STAR*D Study Team. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry2006; 163:28–40.
Balon R, Mufti R, Arfken CL. A survey of prescribing practices for monoamine oxidase inhibitors. Psychiatr Serv1999; 50:945–947.
Clary C, Mandos LA, Schweizer E. Results of a brief survey on the prescribing practices for monoamine oxidase inhibitor antidepressants. J Clin Psychiatry1990; 51:226–231.
Shulman KI, Fischer HD, Herrmann N, Huo CY, Anderson GM, Rochon PA. Current prescription patterns and safety profile of irreversible monoamine oxidase inhibitors: a population-based cohort study of older adults. J Clin Psychiatry2009; 70:1681–1686.
Horwitz D, Lovenberg W, Engelman K, Sjoerdsma A. Monoamine oxidase inhibitors, tyramine, and cheese. JAMA1964; 188:1108–1110.
Asatoor AM, Levi AJ, Milne MD. Tranylcypromine and cheese. Lancet1963; 2:733–734.
Gordon MN, Muller CD, Sherman KA, Morgan DG, Azzaro AJ, Wecker L. Oral versus transdermal selegiline: antidepressant-like activity in rats. Pharmacol Biochem Behav1999; 63:501–506.
Lotufu-Neto F, Trivedi M, Thase ME. Meta-analysis of the reversible inhibitors of monoamine oxidase type A moclobemide and brofaromine for the treatment of depression. Neuropsychopharmacology1999; 20:226–247.
Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract2004; 10:239–248.
Evans DL, Davidson J, Raft D. Early and late side effects of phenelzine. J Clin Psychopharmacol1982; 2:208–210.
Fava M. Weight gain and antidepressants. J Clin Psychiatry2000; 61(suppl 11):37–41.
Rabkin J, Quitkin F, Harrison W, Tricamo E, McGrath P. Adverse reactions to monoamine oxidase inhibitors. Part I. A comparative study. J Clin Psychopharmacol1984; 4:270–278.
Gomez-Gil E, Salmeron JM, Mas A. Phenelzine-induced fulminant hepatic failure. Ann Intern Med1996; 124:692–693.
Lavin MR, Mendelwitz A, Kronig MH. Spontaneous hypertensive reactions with monoamine oxidase inhibitors. Biol Psychiatry1993; 34:146–151.
Fisher P. Serotonin syndrome in the elderly after antidepressive monotherapy. J Clin Psychopharmacol1995; 15:440–442.
Sternback H. The serotonin syndrome. Am J Psychiatry1991; 148:705–713.
Thase M. Novel transdermal delivery formulation of the monoamine oxidase inhibitor selegiline nearing release for treatment of depression. J Clin Psychiatry2006; 67:671–672.
Lee KC, Chen JJ. Transdermal selegiline for the treatment of major depressive disorder. Neuropsychiatr Dis Treat2007; 3:527–537.
Cooper AJ. Tyramine and irreversible monoamine oxidase inhibitors in clinical practice. Br J Psychiatry Suppl1989; Oct(6):38–45.
Mallinger AG, Smith E. Pharmacokinetics of monoamine oxidase inhibitors. Psychopharmacol Bull1991; 27:493–502.
Fulton B, Benfield P. Moclobemide. An update of its pharmacological properties and therapeutic use. Drugs1996; 52:450–474.
Marangell LB. Switching antidepressants for treatment-resistant major depression. J Clin Psychiatry2001; 62(suppl 18):12–17.
Gitlin MJ. Venlafaxine, monoamine oxidase inhibitors, and the serotonin syndrome. J Clin Psychopharmacol1997; 17:66–67.
Szuba MP, Hornig-Rohan M, Amsterdam JD. Rapid conversion from one monoamine oxidase inhibitor to another. J Clin Psychiatry1997; 58:307–310.
Lorenz RA, Vandenberg AM, Canepa EA. Serotonergic antidepressants and linezolid: a retrospective chart review and presentation of cases. Int J Psychiatry Med2008; 38:81–90.
Miller DG, Lovell EO. Antibiotic-induced serotonin syndrome. J Emerg Med2008, May1(Epub ahead of print).
Das PK, Wakentin DI, Hewko R, Forrest DL. Serotonin syndrome after concomitant treatment with linezolid and meperidine. Clin Infect Dis2008; 46:264–265.
Packer S, Berman SA. Serotonin syndrome precipitated by the monoamine oxidase inhibitor linezolid. Am J Psychiatry2007; 164:346–347.
Diamond S. The use of sumatriptan in patients on monoamine oxidase inhibitors. Neurology1995; 45:1039–1040.
Fox AW. Subcutaneous sumatriptan pharmacokinetics: delimiting the monoamine oxidase inhibitor effect. Headache2010; 50:249–255.
Gardner DM, Shulman KI, Walker SE, Tailor SA. The making of a user friendly MAOI diet. J Clin Psychiatry1996; 57:99–104.
Sweet RA, Brown EJ, Heimberg RG, et al. Monoamine oxidase inhibitor dietary restrictions: what are we asking patients to give up?J Clin Psychiatry1995; 56:196–201.
Shulman KI, Walker SE. Refining the MAOI diet: tyramine content of pizzas and soy products. J Clin Psychiatry1999; 60:191–193.
Azzaro AJ, Vandenberg CM, Blob LF, et al. Tyramine pressor sensitivity during treatment with the selegiline transdermal system 6 mg/24 h in healthy subjects. J Clin Pharmacol2006; 46:933–944.
Quitkin FM, Stewart JW, McGrath PJ, et al. Columbia atypical depression. A subgroup of depressives with better response to MAOI than to tricyclic antidepressants or placebo. Br J Psychiatry Suppl1993; Sep(21):30–34.
Davidson J, Pelton S. Forms of atypical depression and their response to antidepressant drugs. Psychiatry Res1986; 17:87–95.
Liebowitz MR, Quitkin FM, Stewart JW, et al. Antidepressant specificity in atypical depression. Arch Gen Psychiatry1988; 45:129–137.
Rapaport MH, Thase ME. Translating the evidence on atypical depression into clinical practice. J Clin Psychiatry2007; 68:e11.
Liebowitz MR. Depression with anxiety and atypical depression. J Clin Psychiatry1993; 54(suppl):10–14.
Stewart JW, McGrath PJ, Quitkin FM, et al. Chronic depression: response to placebo, imipramine, and phenelzine. J Clin Psychopharmacol1993; 13:391–396.
McGrath PJ, Stewart JW, Nunes EV, et al. A double-blind crossover trial of imipramine and phenelzine for outpatients with treatment-refractory depression. Am J Psychiatry1993; 150:118–123.
Zarate CA, Tohen M, Baraibar G, Kando JC, Mirin J. Prescribing trends of antidepressants in bipolar depression. J Clin Psychiatry1995; 56:260–264.
Mallinger AG, Frank E, Thase ME, Barwell MM, Diazgranados N, Luckenbaugh DA, Kupfer DJ. Revisiting the effectiveness of standard antidepressants in bipolar disorder: are monoamine oxidase inhibitors superior?Psychopharmacol Bull2009; 42:64–74.
Anderson IM, Nutt DJ, Deakin JF. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 1993 British Association for Psychopharmacology guidelines. British Association for Psychopharmacology. J Psychopharmacol2000; 14:3–20.
Liebowitz MR, Hollander E, Schneier F, et al. Reversible and irreversible monoamine oxidase inhibitors in other psychiatric disorders. Acta Psychiatr Scand Suppl1990; 360:29–34.
Davidson JR, Giller EL, Zisook S, Overall JE. An efficacy study of isocarboxazid and placebo in depression, and its relationship to depressive nosology. Arch Gen Psychiatry1988; 45:120–127.
Razani J, White KL, White J, et al. The safety and efficacy of combined amitriptyline and tranylcypromine antidepressant treatment: a controlled trial. Arch Gen Psychiatry1983; 40:657–661.
Thase ME, Triverdi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology1995; 12:185–219.
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision. Washington, DC, American Psychiatric Association, 2000.
Parker G, Crawford J. Atypical depression: retrospective self-reporting of treatment effectiveness. Acta Psychiatr Scand2009; 120:213–221.
Henkel V, Mergl R, Algaier AK, Kohnen R, Moller HJ, Hergerl U. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res2006; 141:89–101.
Rothschild R, Quitkin HM, Quitkin FM, et al. A double-blind placebo-controlled comparison of phenelzine and imipramine in the treatment of bulimia in atypical depressives. Int J Eat Disord1994; 15:1–9.
Judd LL, Paulus MJ, Schettler PJ, et al. Does incomplete recovery from first life-time major depressive episode herald a chronic course of illness?Am J Psychiatry2000; 157:1501–1504.
Murphy JM, Monson RR, Olivier DC, Sobol AM, Leighton AH. Affective disorders and mortality: a general population study. Arch Gen Psychiatry1987; 44:473–480.
Thase ME, Frank E, Mallinger AG, Hamer T, Kupfer DJ. Treatment of imipramine-resistant recurrent depression, III: efficacy of monoamine oxidase inhibitors. J Clin Psychiatry1992; 53:5–11.
Thase ME, Mallinger AG, McKnight D, Himmelhoch JM. Treatment of imipramine-resistant recurrent depression, IV: a double blind crossover study of tranylcypromine for anergic bipolar depression. Am J Psychiatry1992; 149:195–198.
Quitkin FM, McGrath PJ, Stewart JW, et al. Atypical depression, panic attacks, and response to imipramine and phenelzine: a replication. Arch Gen Psychiatry1990; 47:935–941.
Himmelhoch JM, Thase ME, Mallinger AG, Houck P. Tranylcypromine versus imipramine in anergic bipolar depression. Am J Psychiatry1991; 148:910–916.
Bodkin JA, Amsterdam JD. Transdermal selegiline in major depression: a double-blind, placebo-controlled, parallel-group study in outpatients. Am J Psychiatry2002; 159:1869–1875.
Amsterdam JD. A double-blind, placebo-controlled trial of the safety and efficacy of selegiline transdermal system without dietary restrictions in patients with major depressive disorder. J Clin Psychiatry2003; 64:208–214.
Amsterdam JD, Bodkin JA. Selegiline transdermal system in the prevention of relapse of major depressive disorder: a 52-week, double-blind, placebo-substitution, parallel-group clinical trial. J Clin Psychopharmacol2006; 26:579–586.
Feiger AD, Rickels K, Rynn MA, et al. Selegiline transdermal system for the treatment of major depressive disorder: an 8-week, double-blind, placebo-controlled, flexible-dose titration trial. J Clin Psychiatry2006; 67:1354–1361.
Data from multiple studies suggest the efficacy of MAO inhibitors in the management of major depressive disorder and, in particular, major depressive disorder with atypical features and in treatment-resistant depression.
When using oral MAO inhibitors, patients must follow a low-tyramine diet to avoid the “cheese reaction,” ie, tyramine-induced hypertensive crisis. However, recent studies suggest that traditional dietary advice may be unnecessarily restrictive.
The selegiline transdermal system (Emsam) is the first approved transdermal patch for treatment of major depression. Unlike oral MAO inhibitors, the patch can be used without the dietary restrictions at its lowest effective dose of 6 mg/24 hours. Because of its transdermal delivery, it has the advantage of not inhibiting the metabolism of dietary tyramine by MAO subtype A in the gut, while providing antidepressant effect in the brain. The patch may be a promising alternative to existing strategies for the management of major depressive disorder.
New clinical trials and observational studies are shedding light on ways to improve the health of elderly patients. Here is a brief summary of these trials and how they might influence your clinical practice.
EXERCISE HAS NEWLY DISCOVERED BENEFITS
According to government data,1 exercise has a dose-dependent effect on rates of all-cause mortality: the more hours one exercises per week, the lower the risk of death. The difference in risk is most pronounced as one goes from no exercise to about 3 hours of exercise per week; above 3 hours per week, the curve flattens out but continues to decline. Hence, we advise patients to engage in about 30 minutes of moderate-intensity exercise every day.
Lately, physical exercise has been found to have other, unexpected benefits.
Exercise helps cognition
ERICKSON KI, PRAKASH RS, VOSS MW, ET AL. AEROBIC FITNESS IS ASSOCIATED WITH HIPPOCAMPAL VOLUME IN ELDERLY HUMANS. HIPPOCAMPUS 2009; 19:1030–1039.
ETGEN T, SANDER D, HUNTGEBURTH U, POPPERT H, FÖRSTL H, BICKEL H. PHYSICAL ACTIVITY AND INCIDENT COGNITIVE IMPAIRMENT IN ELDERLY PERSONS: THE INVADE STUDY. ARCH INTERN MED 2010; 170:186–193.
The hippocampus is a structure deep in the brain that is involved in short-term memory. It atrophies with age, more so with dementia. Erickson2 found a correlation between aerobic fitness (as measured by maximum oxygen consumption), hippocampal volume, and spatial memory performance.
Etgen and colleagues3 studied nearly 4,000 older adults in Bavaria for 2 years. Among those reporting no physical activity, 21.4% had cognitive impairment at baseline, compared with 7.3% of those with high activity at baseline. Following those without cognitive impairment over a 2-year period, they found the incidence of new cognitive impairment was 13.9% in those with no physical activity at baseline, 6.7% in those with moderate activity, and 5.1% in those with high activity.
Exercise boosts the effect of influenza vaccine
WOODS JA, KEYLOCK KT, LOWDER T, ET AL. CARDIOVASCULAR EXERCISE TRAINING EXTENDS INFLUENZA VACCINE SEROPROTECTION IN SEDENTARY OLDER ADULTS: THE IMMUNE FUNCTION INTERVENTION TRIAL. J AM GERIATR SOC 2009; 57:2183–2191.
In a study in 144 sedentary but healthy older adults (ages 60 to 83), Woods et al4 randomized the participants to undergo either flexibility or cardiovascular training for 10 months, starting 4 months before their annual influenza shot. Exercise extended the duration of antibody protection, with more participants in the cardiovascular group than in the flexibility group showing protection at 24 weeks against all three strains covered by the vaccine: H1N1, H3N2, and influenza B.
PREVENTING FRACTURES
Each year, about 30% of people age 65 or older fall, sustaining serious injuries in 5% to 10% of cases. Unintentional falls are the main cause of hip fractures, which number 300,000 per year. They are also a common cause of death.
Vitamin D prevents fractures, but can there be too much of a good thing?
BISCHOFF-FERRARI HA, WILLETT WC, WONG JB, ET AL. PREVENTION OF NONVERTEBRAL FRACTURES WITH ORAL VITAMIN D AND DOSE DEPENDENCY: A META-ANALYSIS OF RANDOMIZED CONTROLLED TRIALS. ARCH INTERN MED 2009; 169:551–561.
SANDERS KM, STUART AL, WILLIAMSON EJ, ET AL. ANNUAL HIGH-DOSE ORAL VITAMIN D AND FALLS AND FRACTURES IN OLDER WOMEN: A RANDOMIZED CONTROLLED TRIAL. JAMA 2010; 303:1815–1822.
Bischoff-Ferrari5 performed a meta-analysis of 12 randomized controlled trials of oral supplemental vitamin D3 for preventing nonvertebral fractures in people age 65 and older, and eight trials for preventing hip fractures in the same age group. They found that the higher the daily dose of vitamin D, the lower the relative risk of hip fracture. The threshold dose at which supplementation significantly reduced the risk of falling was about 400 units per day. Higher doses of vitamin D reduced both falls and hip fractures by about 20%. The maximal effect was seen with studies using the maximum daily doses, ie, 770 to 800 units per day—not megadoses, but more than most Americans are taking. The threshold serum level of vitamin D of significance was 60 nmol/L (24 ng/mL).
Of interest, the effect on fractures was independent of calcium supplementation. This is important because calcium supplementation over and above ordinary dietary intake may increase the risk of cardiovascular events.6,7
Despite the benefits of vitamin D, too much may be too much of a good thing. Sanders et al8 performed a double-blind, placebo-controlled trial in 2,256 community-dwelling women, age 70 or older, who were considered to be at high risk for fractures. Half received a large oral dose (500,000 units) once a year for 3 to 5 years, and half got placebo. Their initial serum vitamin D level was 49 nmol/L; the level 30 days after a dose in the treatment group was 120 nmol/L.
Contrary to expectations, the incidence of falls was 15% higher in the vitamin D group than in the placebo group (P = .03), and the incidence of fractures was 26% higher (P = .047). The falls and fractures tended to cluster in the first 3 months after the dose in the active treatment group, when serum vitamin D levels were highest.
Comments. Unless future studies suggest a benefit to megadoses of vitamin D or prove calcium supplementation greater than 1,000 mg is safe, the optimal daily intake of vitamin D is likely 1,000 units, with approximately 200 units from diet and 800 units from supplements. A diet rich in low-fat dairy products may not require calcium supplementation. In those consuming a low-calcium diet, supplements of 500 to 1,000 mg/day are likely adequate.
Denosumab, a new drug for preventing fractures
CUMMINGS SR, SAN MARTIN J, MCCLUNG MR, ET AL; FREEDOM TRIAL. DENOSUMAB FOR PREVENTION OF FRACTURES IN POSTMENOPAUSAL WOMEN WITH OSTEOPOROSIS. N ENGL J MED 2009; 361:756–765.
SMITH MR, EGERDIE B, HERNÁNDEZ TORIZ N, ET AL; DENOSUMAB HALT PROSTATE CANCER STUDY GROUP. DENOSUMAB IN MEN RECEIVING ANDROGEN-DEPRIVATION THERAPY FOR PROSTATE CANCER. N ENGL J MED 2009; 361:745–755.
Denosumab (Prolia) is the first of a new class of drugs for the treatment of osteoporosis. It is a monoclonal antibody and member of the tumor necrosis factor superfamily that binds to the receptor activator nuclear factor kappa B (RANK) ligand. It has an antiresorptive effect, preventing osteoclast differentiation and activation. It is given by subcutaneous injection of 60 mg every 6 months; it is cleared by a nonrenal mechanism.
In a randomized controlled trial in 7,868 women between the ages of 60 and 90 who had osteoporosis, Cummings et al9 reported that denosumab reduced the 3-year incidence of vertebral fractures by 68% (P < .001), reduced the incidence of hip fractures by 40% (P = .01), and reduced the incidence of nonvertebral fractures by 20% (P = .01). In a trial in men receiving androgen deprivation therapy for prostate cancer, Smith et al10 reported that denosumab reduced the incidence of vertebral fracture by 62% (P = .006).
Comment. Denosumab was approved by the US Food and Drug Administration (FDA) on June 1, 2010, and is emerging in specialty clinics at the time of this publication. Its potential impact on clinical care is not yet known. It is costly—about $825 (average wholesale price) per injection—but since it is given by injection it may be easier than a yearly infusion of zoledronic acid (Reclast). It has the potential to suppress immune function, although this was not reported in the clinical trials. It may ultimately have a role in treating osteoporosis in men and women, prostate cancer following androgen deprivation, metastatic prostate cancer, metastatic breast cancer, osteoporosis with renal impairment, and other diseases.
DIALYSIS IN THE ELDERLY: A BLEAK STORY
KURELLA TAMURA M, COVINSKY KE, CHERTOW GM, YAFFE K, LANDEFELD CS, MCCOLLOCH CE. FUNCTIONAL STATUS OF ELDERLY ADULTS BEFORE AND AFTER INITIATION OF DIALYSIS. N ENGL J MED 2009; 361:1539–1547.
JASSAL SV, CHIU E, HLADUNEWITH M. LOSS OF INDEPENDENCE IN PATIENTS STARTING DIALYSIS AT 80 YEARS OF AGE OR OLDER (LETTER). N ENGL J MED 2009; 361:1612–1613.
Nursing home residents account for 4% of all patients in end-stage renal disease. However, the benefits of dialysis in older patients are uncertain. The mortality rate during the first year of dialysis is 35% in patients 70 years of age and older and 50% in patients 80 years and older.
Is dialysis helpful in the elderly, ie, does it improve survival and function?
Kurella Tamura et al11 retrospectively identified 3,702 nursing home residents starting dialysis in whom functional assessments had been done. The numbers told a bleak story. Initiation of dialysis was associated with a sharp decline in functional status, as reflected in an increase of 2.8 points on the 28-point Minimum Data Set–Activities of Daily Living (MDS-ADL) scale (the higher the score, the worse the function). MDS-ADL scores stabilized at a plateau for about 6 months and then continued to decline. Moreover, at 12 months, 58% of the patients had died.
The MDS-ADL score is based on seven components: eating, bed mobility, locomotion, transferring, toileting, hygiene, and dressing; function declined in all of these areas when patients started dialysis.
Patients were more likely to decline in activities of daily living after starting dialysis if they were older, were white, had cerebrovascular disease, had a diagnosis of dementia, were hospitalized at the start of dialysis, or had a serum albumin level lower than 3.5 g/dL.
The same thing happens to elders living in the community when they start dialysis. Jassal and colleagues12 reported that, of 97 community-dwelling patients (mean age 85), 46 (47%) were dead 2 years after starting dialysis. Although 76 (78%) had been living independently at the start of dialysis, only 11 (11%) were still doing so at 2 years.
Comment. These findings indicate that we do not know if hemodialysis improves survival. Hemodialysis may buy about 3 months of stable function, but it clearly does not restore function.
Is this the best we can do? Standard hemodialysis may have flaws, and nocturnal dialysis and peritoneal dialysis are used more in other countries. These dialysis techniques require more study in our older population. The lesson from these two publications on dialysis is that we should attend more carefully to slowing the decline in renal function before patients reach end-stage renal disease.
DABIGATRAN: AN ALTERNATIVE TO WARFARIN FOR ATRIAL FIBRILLATION
CONNOLLY SJ, EZEKOWITZ MD, YUSUF S, ET AL; RE-LY STEERING COMMITTEE AND INVESTIGATORS. DABIGATRAN VERSUS WARFARIN IN PATIENTS WITH ATRIAL FIBRILLATION. N ENGL J MED 2009; 361:1139–1151.
Atrial fibrillation is common, affecting 2.2 million adults. The median age of people who have atrial fibrillation is 75 years, and it is the most common arrhythmia in the elderly. Some 20% of ischemic strokes are attributed to it.13–15
Warfarin (Coumadin) is still the mainstay of treatment to prevent stroke in patients with atrial fibrillation. In an analysis of pooled data from five clinical trials,16 the relative risk reduction with warfarin was about 68% in the overall population (number needed to treat 32), 51% in people older than 75 years with no other risk factors (number needed to treat 56), and 85% in people older than 75 years with one or more risk factors (number needed to treat 15).
But warfarin carries a risk of bleeding, and its dose must be periodically adjusted on the basis of the international normalized ratio (INR) of the prothrombin time, so it carries a burden of laboratory monitoring. It is less safe in people who eat erratically, resulting in wide fluctuations in the INR.
Dabigatran (Pradaxa), a direct thrombin inhibitor, is expected to become an alternative to warfarin. It has been approved in Europe but not yet in the United States.
Connolly et al,17 in a randomized, double-blind trial, assigned 18,113 patients who had atrial fibrillation to receive either dabigatran 110 or 150 mg twice daily or adjusted-dose warfarin in an unblinded fashion. At 2 years, the rates of stroke and systemic embolism were about the same with dabigatran 110 mg as with warfarin but were lower with dabigatran 150 mg (relative risk 0.66, 95% confidence interval [CI] 0.53–0.82, P < .001). The rate of major bleeding was lower with dabigatran 110 mg than with warfarin (2.71% per year vs 3.36% per year, P = .003), but it was similar with dabigatran 150 mg (3.11% per year). Rates of life-threatening bleeding were 1.80% with warfarin, 1.22% with dabigatran 110 mg (P < .05), and 1.45% with dabigatran 150 mg (P < .05).
Comment. I suspect that warfarin’s days are numbered. Dabigatran 110 or 150 mg was as safe and as effective as warfarin in clinical trials, and probably will be more effective than warfarin in clinical practice. It will also probably be safer than warfarin in clinical practice, particularly in challenging settings such as long-term care. On the other hand, it will likely be much more expensive than warfarin.
DEMENTIA
Adverse effects of cholinesterase inhibitors
GILL SS, ANDERSON GM, FISCHER HD, ET AL. SYNCOPE AND ITS CONSEQUENCES IN PATIENTS WITH DEMENTIA RECEIVING CHOLINESTERASE INHIBITORS: A POPULATION-BASED COHORT STUDY. ARCH INTERN MED 2009; 169:867–873.
Cholinesterase inhibitors, eg, donepezil (Aricept), galantamine (Razadyne), and rivastigmine (Exelon), are commonly used to treat Alzheimer disease. However, these drugs carry risks of serious adverse effects.
Gill et al18 retrospectively reviewed a database from Ontario, Canada, and identified about 20,000 community-dwelling elderly persons admitted to the hospital who had been prescribed cholinesterase inhibitors and about three times as many matched controls.
Several adverse events were more frequent in people receiving cholinesterase inhibitors. Findings (events per 1,000 person-years):
Hospital visits for syncope: 31.5 vs 18.6, adjusted hazard ratio (HR) 1.76, 95% CI 1.57–1.98
Hip fractures: 22.4 vs 19.8, HR 1.18, 85% CI 1.04–1.34
Hospital visits for bradycardia: 6.9 vs 4.4, HR 1.69, 95% CI 1.32–2.15
Permanent pacemaker insertion: 4.7 vs 3.3, HR 1.49, 95% CI 1.12–2.00.
Comment. This study adds to the concerns that cholinesterase inhibitors, which have only modest cognitive benefits, may increase the risk of falls, injury, and need for pacemaker placement in demented patients. A low threshold to stop medications in this class should be considered when a patient on a cholinesterase inhibitor presents with bradycardia, falls, and syncope.
The importance of ‘staging’ dementia
IVERSON DJ, GRONSETH GS, REGER MA, ET AL; STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. PRACTICE PARAMETER UPDATE: EVALUATION AND MANAGEMENT OF DRIVING RISK IN DEMENTIA: REPORT OF THE QUALITY STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. NEUROLOGY 2010; 74:1316–1324.
The Clinical Dementia Rating (CDR) is a simple scale that should be applied by clinicians to describe stage of dementia in patients with Alzheimer disease. This scale can be useful in a variety of settings, from prescribing antidementia drugs to determining whether a patient should still drive. Although research protocols utilize a survey or semistructured interview to derive the stage, the clinician can estimate the stage easily in the office, particularly if there is an informant who can comment on performance outside the office.
There are four stages to the CDR19:
0: No dementia
0.5: Mild memory deficit but intact function
1.0: Moderate memory loss with mild functional impairment
2.0: Severe memory loss, moderate functional impairment
3.0: Severe memory loss, no significant function outside of the house.
Comment. The first stage (0.5, mild memory deficit but intact function) corresponds to “mild cognitive impairment.” In the clinic, these patients tend to take more notes. They come to the appointment with a little book and they write everything down so they don’t forget. They do arrive at their appointments on time; they are not crashing the car; they are paying their bills.
Patients with CDR stage 1.0 dementia (moderate memory loss with mild functional impairment) may miss appointments, they may confuse their medications, and they may have problems driving. They are still taking care of their basic needs, and they show up for appointments acceptably washed and dressed. However, they are likely having trouble shopping and managing their finances.
Patients with severe memory loss and moderate functional impairment (CDR stage 2.0) may not realize they haven’t bathed for a week or have worn the same clothes repeatedly. They are having trouble with basic activities of daily living, such as bathing and toilet hygiene. However, if you were to encounter them socially and didn’t talk to them for too long, you might think they were normal.
Those with severe memory loss and no significant function outside the house (CDR stage 3.0) are the most severely disabled. Dementia in these individuals is recognizable at a glance, from across the room.
Alzheimer patients progress through the stages, from CDR stage 0.5 at about 1 year to stage 1 by about 2 years, to stage 2 by 5 years, and to stage 3 at 8 or 9 years.20
In prescribing antidementia medications. The CDR can help with prescribing antidementia drugs. No medications are approved by the FDA for stage 0 or 0.5. Cholinesterase inhibitors are approved for stages 1, 2, and 3; memantine (Namenda) is approved for stages 2 and 3.
Advising about driving. The CDR is the only risk predictor with a quality-of-evidence rating of A. More than half of people with stage 0.5 memory impairment are safe drivers; fewer than half of those with stage 1.0 are still safe drivers; and patients with stage 2.0 dementia should not be driving at all.21 An adverse rating by a caregiver carries a quality-of-evidence rating of B. Predictors of driving risk with a quality-of-evidence rating of C are decreased mileage due to self-restriction, agitation, or aggression; a crash in the past 1 to 5 years; a citation in the past 2 to 3 years; and a Folstein Mini-Mental State Examination score of 24 or less. Studies also show that a memory-impaired person’s self-rating of safe driving ability or of assurance that he or she avoids unsafe situations is not reliable.21
DELIRIUM
Delirium goes by a number of synonyms, eg, “sundowning,” acute confusional state, acute change in mental status, metabolic encephalopathy, toxic encephalopathy (psychosis), acute brain syndrome, and acute toxic psychosis.
Delirium is common in hospitalized elderly patients, occurring in 11% to 42% of elderly hospitalized patients overall, up to 53% of elderly surgical patients on regular hospital floors, 80% of elderly surgical patients in intensive care, and about half of elderly patients after undergoing coronary artery bypass grafting. Unfortunately, it is undiagnosed in 30% to 60% of cases.22–24
Many pathways can lead to delirium, including hypoxemia, metabolic derangement, drug effects, systemic inflammation, and infection.25
Outcomes can vary from full recovery to death. After 1 year, 50% of those who leave the hospital with some evidence of delirium have not regained their baseline function. Delirium also increases the cost of care and the risk of institutionalization.
Delirium can accelerate dementia
FONG TG, JONES RN, SHI P, ET AL. DELIRIUM ACCELERATES COGNITIVE DECLINE IN ALZHEIMER DISEASE. NEUROLOGY 2009; 72:1570–1575.
Delirium accelerates the course of dementia in patients who had some evidence of dementia before they entered the hospital. Often, the change is noticeable by the family.26
Preventing delirium
INOUYE SK BOGARDUS ST JR, CHARPENTIER PA, ET AL. A MULTICOMPONENT INTERVENTION TO PREVENT DELIRIUM IN HOSPITALIZED OLDER PATIENTS. N ENGL J MED 1999; 340:669–676.
LUNDSTRÖM M, OLOFSSON B, STENVALL M, ET AL. POSTOPERATIVE DELIRIUM IN OLD PATIENTS WITH FEMORAL NECK FRACTURE: A RANDOMIZED INTERVENTION STUDY. AGING CLIN EXP RES 2007; 19:178–186.
Delirium can often be prevented. In a report published in 1999, Inouye et al27 described the outcomes of a program to prevent delirium in hospitalized medically ill elderly patients. Interventions were aimed at optimizing cognitive function, preventing sleep deprivation, avoiding immobility, improving vision and hearing, and treating dehydration. The incidence of delirium was 9.9% in the intervention group vs 15% in the control group, a 40% reduction (P < .05).
Lundström et al28 implemented a similar program for elderly patients with hip fractures. Interventions included staff education and teamwork; active prevention, detection, and treatment of delirium; transfusions if hemoglobin levels were less than 10 g/dL; prompt removal of indwelling urinary catheters, with screening for urinary retention; active prevention and treatment of constipation; and protein-enriched meals. The incidence of delirium was 55% in the intervention group vs 75% in the control group, a 27% reduction.
Comment. Although we have long known that the risk of delirium in medical and surgical patients can be reduced, most hospitals do not have systematic programs to detect delirium and reduce its incidence. Hopefully, reduction in delirium risk will also reduce its adverse consequences, including worsening of dementia and increased mortality.
Erickson KI, Prakash RS, Voss MW, et al. Aerobic fitness is associated with hippocampal volume in elderly humans. Hippocampus2009; 19:1030–1039.
Etgen T, Sander D, Huntgeburth U, Poppert H, Förstl H, Bickel H. Physical activity and incident cognitive impairment in elderly persons: the INVADE study. Arch Intern Med2010; 170:186–193.
Woods JA, Keylock KT, Lowder T, et al. Cardiovascular exercise training extends influenza vaccine seroprotection in sedentary older adults: the immune function intervention trial. J Am Geriatr Soc2009; 57:2183–2191.
Bischoff-Ferrari HA, Willett WC, Wong JB, et al. Prevention of nonvertebral fractures with oral vitamin D and dose dependency: a meta-analysis of randomized controlled trials. Arch Intern Med2009; 169:551–561.
Bolland MJ, Avenell A, Baron JA, et al. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. BMJ2010; 341:c3691. doi:10.1136/bmj.c3691.
Bolland MJ, Barber PA, Doughty RN, et al. Vascular events in healthy older women receiving calcium supplementation: randomised controlled trial. BMJ2008; 336:262–266.
Sanders KM, Stuart AL, Williamson EJ, et al. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA2010; 303:1815–1822.
Cummings SR, San Martin J, McClung MR, et al; FREEDOM Trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med2009; 361:756–765.
Smith MR, Egerdie B, Hernández Toriz N, et al; Denosumab HALT Prostate Cancer Study Group. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med2009; 361:745–755.
Kurella Tamura M, Covinsky KE, Chertow GM, Yaffe K, Landefeld CS, McColloch CE. Functional status of elderly adults before and after initiation of dialysis. N Engl J Med2009; 361:1539–1547.
Jassal SV, Chiu E, Hladunewich M. Loss of independence in patients starting dialysis at 80 years of age or older (letter). N Engl J Med2009; 361:1612–1613.
Feinberg WM, Blackshear JL, Laupacis A, Kronmal R, Hart RG. Prevalence, age distribution and gender of patients with atrial fibrillation. Analysis and implications. Arch Intern Med1995; 155:469–473.
Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation: a major contributor to stroke in the elderly. The Framingham Study. Arch Intern Med1987; 147:1561–1564.
Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial fibrillation. The Framingham Study. Stroke1996; 27:1760–1764.
Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med1994; 154:1449–1457.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern Med2009; 169:867–873.
Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology1993; 43:2412–2414.
Sloane PD. Advances in the treatment of Alzheimer’s disease. Am Fam Physician1998; 58:1577–1586.
Iverson DJ, Gronseth GS, Reger MA, et al; Standards Subcommittee of the American Academy of Neurology. Practice parameter update: evaluation and management of driving risk in dementia: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology2010; 74:1316–1324.
Demeure MJ, Fain MJ. The elderly surgical patient and postoperative delirium. J Am Coll Surg2006; 203:752–757.
Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing2006; 35:350–364.
Rudolph JL, Jones RN, Levkoff SE, et al. Derivation and validation of a preoperative prediction rule for delirium after cardiac surgery. Circulation2009; 119:229–236.
Fong TG, Tulebaev SR, Inouye SK. Delirium in elderly adults: diagnosis, prevention and treatment. Nat Rev Neurol2009; 5:210–220.
Fong TG, Jones RN, Shi P, et al. Delirium accelerates cognitive decline in Alzheimer disease. Neurology2009; 72:1570–1575.
Inouye SK, Bogardus ST, Charpentier PA, et al. A multicomponent intervention to prevent delirium in hospitalized older patients. N Engl J Med1999; 340:669–676.
Lundström M, Olofsson B, Stenvall M, et al. Postoperative delirium in old patients with femoral neck fracture: a randomized intervention study. Aging Clin Exp Res2007; 19:178–186.
Barbara Messinger-Rapport, MD, PhD, CMD Director, Center for Geriatric Medicine, Medicine Institute, Cleveland Clinic
Address: Barbara M. Messinger-Rapport, MD, PhD, Center for Geriatric Medicine, G10, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Medical Grand Rounds articles are based on edited transcripts from Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.
Barbara Messinger-Rapport, MD, PhD, CMD Director, Center for Geriatric Medicine, Medicine Institute, Cleveland Clinic
Address: Barbara M. Messinger-Rapport, MD, PhD, Center for Geriatric Medicine, G10, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Medical Grand Rounds articles are based on edited transcripts from Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.
Author and Disclosure Information
Barbara Messinger-Rapport, MD, PhD, CMD Director, Center for Geriatric Medicine, Medicine Institute, Cleveland Clinic
Address: Barbara M. Messinger-Rapport, MD, PhD, Center for Geriatric Medicine, G10, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Medical Grand Rounds articles are based on edited transcripts from Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.
New clinical trials and observational studies are shedding light on ways to improve the health of elderly patients. Here is a brief summary of these trials and how they might influence your clinical practice.
EXERCISE HAS NEWLY DISCOVERED BENEFITS
According to government data,1 exercise has a dose-dependent effect on rates of all-cause mortality: the more hours one exercises per week, the lower the risk of death. The difference in risk is most pronounced as one goes from no exercise to about 3 hours of exercise per week; above 3 hours per week, the curve flattens out but continues to decline. Hence, we advise patients to engage in about 30 minutes of moderate-intensity exercise every day.
Lately, physical exercise has been found to have other, unexpected benefits.
Exercise helps cognition
ERICKSON KI, PRAKASH RS, VOSS MW, ET AL. AEROBIC FITNESS IS ASSOCIATED WITH HIPPOCAMPAL VOLUME IN ELDERLY HUMANS. HIPPOCAMPUS 2009; 19:1030–1039.
ETGEN T, SANDER D, HUNTGEBURTH U, POPPERT H, FÖRSTL H, BICKEL H. PHYSICAL ACTIVITY AND INCIDENT COGNITIVE IMPAIRMENT IN ELDERLY PERSONS: THE INVADE STUDY. ARCH INTERN MED 2010; 170:186–193.
The hippocampus is a structure deep in the brain that is involved in short-term memory. It atrophies with age, more so with dementia. Erickson2 found a correlation between aerobic fitness (as measured by maximum oxygen consumption), hippocampal volume, and spatial memory performance.
Etgen and colleagues3 studied nearly 4,000 older adults in Bavaria for 2 years. Among those reporting no physical activity, 21.4% had cognitive impairment at baseline, compared with 7.3% of those with high activity at baseline. Following those without cognitive impairment over a 2-year period, they found the incidence of new cognitive impairment was 13.9% in those with no physical activity at baseline, 6.7% in those with moderate activity, and 5.1% in those with high activity.
Exercise boosts the effect of influenza vaccine
WOODS JA, KEYLOCK KT, LOWDER T, ET AL. CARDIOVASCULAR EXERCISE TRAINING EXTENDS INFLUENZA VACCINE SEROPROTECTION IN SEDENTARY OLDER ADULTS: THE IMMUNE FUNCTION INTERVENTION TRIAL. J AM GERIATR SOC 2009; 57:2183–2191.
In a study in 144 sedentary but healthy older adults (ages 60 to 83), Woods et al4 randomized the participants to undergo either flexibility or cardiovascular training for 10 months, starting 4 months before their annual influenza shot. Exercise extended the duration of antibody protection, with more participants in the cardiovascular group than in the flexibility group showing protection at 24 weeks against all three strains covered by the vaccine: H1N1, H3N2, and influenza B.
PREVENTING FRACTURES
Each year, about 30% of people age 65 or older fall, sustaining serious injuries in 5% to 10% of cases. Unintentional falls are the main cause of hip fractures, which number 300,000 per year. They are also a common cause of death.
Vitamin D prevents fractures, but can there be too much of a good thing?
BISCHOFF-FERRARI HA, WILLETT WC, WONG JB, ET AL. PREVENTION OF NONVERTEBRAL FRACTURES WITH ORAL VITAMIN D AND DOSE DEPENDENCY: A META-ANALYSIS OF RANDOMIZED CONTROLLED TRIALS. ARCH INTERN MED 2009; 169:551–561.
SANDERS KM, STUART AL, WILLIAMSON EJ, ET AL. ANNUAL HIGH-DOSE ORAL VITAMIN D AND FALLS AND FRACTURES IN OLDER WOMEN: A RANDOMIZED CONTROLLED TRIAL. JAMA 2010; 303:1815–1822.
Bischoff-Ferrari5 performed a meta-analysis of 12 randomized controlled trials of oral supplemental vitamin D3 for preventing nonvertebral fractures in people age 65 and older, and eight trials for preventing hip fractures in the same age group. They found that the higher the daily dose of vitamin D, the lower the relative risk of hip fracture. The threshold dose at which supplementation significantly reduced the risk of falling was about 400 units per day. Higher doses of vitamin D reduced both falls and hip fractures by about 20%. The maximal effect was seen with studies using the maximum daily doses, ie, 770 to 800 units per day—not megadoses, but more than most Americans are taking. The threshold serum level of vitamin D of significance was 60 nmol/L (24 ng/mL).
Of interest, the effect on fractures was independent of calcium supplementation. This is important because calcium supplementation over and above ordinary dietary intake may increase the risk of cardiovascular events.6,7
Despite the benefits of vitamin D, too much may be too much of a good thing. Sanders et al8 performed a double-blind, placebo-controlled trial in 2,256 community-dwelling women, age 70 or older, who were considered to be at high risk for fractures. Half received a large oral dose (500,000 units) once a year for 3 to 5 years, and half got placebo. Their initial serum vitamin D level was 49 nmol/L; the level 30 days after a dose in the treatment group was 120 nmol/L.
Contrary to expectations, the incidence of falls was 15% higher in the vitamin D group than in the placebo group (P = .03), and the incidence of fractures was 26% higher (P = .047). The falls and fractures tended to cluster in the first 3 months after the dose in the active treatment group, when serum vitamin D levels were highest.
Comments. Unless future studies suggest a benefit to megadoses of vitamin D or prove calcium supplementation greater than 1,000 mg is safe, the optimal daily intake of vitamin D is likely 1,000 units, with approximately 200 units from diet and 800 units from supplements. A diet rich in low-fat dairy products may not require calcium supplementation. In those consuming a low-calcium diet, supplements of 500 to 1,000 mg/day are likely adequate.
Denosumab, a new drug for preventing fractures
CUMMINGS SR, SAN MARTIN J, MCCLUNG MR, ET AL; FREEDOM TRIAL. DENOSUMAB FOR PREVENTION OF FRACTURES IN POSTMENOPAUSAL WOMEN WITH OSTEOPOROSIS. N ENGL J MED 2009; 361:756–765.
SMITH MR, EGERDIE B, HERNÁNDEZ TORIZ N, ET AL; DENOSUMAB HALT PROSTATE CANCER STUDY GROUP. DENOSUMAB IN MEN RECEIVING ANDROGEN-DEPRIVATION THERAPY FOR PROSTATE CANCER. N ENGL J MED 2009; 361:745–755.
Denosumab (Prolia) is the first of a new class of drugs for the treatment of osteoporosis. It is a monoclonal antibody and member of the tumor necrosis factor superfamily that binds to the receptor activator nuclear factor kappa B (RANK) ligand. It has an antiresorptive effect, preventing osteoclast differentiation and activation. It is given by subcutaneous injection of 60 mg every 6 months; it is cleared by a nonrenal mechanism.
In a randomized controlled trial in 7,868 women between the ages of 60 and 90 who had osteoporosis, Cummings et al9 reported that denosumab reduced the 3-year incidence of vertebral fractures by 68% (P < .001), reduced the incidence of hip fractures by 40% (P = .01), and reduced the incidence of nonvertebral fractures by 20% (P = .01). In a trial in men receiving androgen deprivation therapy for prostate cancer, Smith et al10 reported that denosumab reduced the incidence of vertebral fracture by 62% (P = .006).
Comment. Denosumab was approved by the US Food and Drug Administration (FDA) on June 1, 2010, and is emerging in specialty clinics at the time of this publication. Its potential impact on clinical care is not yet known. It is costly—about $825 (average wholesale price) per injection—but since it is given by injection it may be easier than a yearly infusion of zoledronic acid (Reclast). It has the potential to suppress immune function, although this was not reported in the clinical trials. It may ultimately have a role in treating osteoporosis in men and women, prostate cancer following androgen deprivation, metastatic prostate cancer, metastatic breast cancer, osteoporosis with renal impairment, and other diseases.
DIALYSIS IN THE ELDERLY: A BLEAK STORY
KURELLA TAMURA M, COVINSKY KE, CHERTOW GM, YAFFE K, LANDEFELD CS, MCCOLLOCH CE. FUNCTIONAL STATUS OF ELDERLY ADULTS BEFORE AND AFTER INITIATION OF DIALYSIS. N ENGL J MED 2009; 361:1539–1547.
JASSAL SV, CHIU E, HLADUNEWITH M. LOSS OF INDEPENDENCE IN PATIENTS STARTING DIALYSIS AT 80 YEARS OF AGE OR OLDER (LETTER). N ENGL J MED 2009; 361:1612–1613.
Nursing home residents account for 4% of all patients in end-stage renal disease. However, the benefits of dialysis in older patients are uncertain. The mortality rate during the first year of dialysis is 35% in patients 70 years of age and older and 50% in patients 80 years and older.
Is dialysis helpful in the elderly, ie, does it improve survival and function?
Kurella Tamura et al11 retrospectively identified 3,702 nursing home residents starting dialysis in whom functional assessments had been done. The numbers told a bleak story. Initiation of dialysis was associated with a sharp decline in functional status, as reflected in an increase of 2.8 points on the 28-point Minimum Data Set–Activities of Daily Living (MDS-ADL) scale (the higher the score, the worse the function). MDS-ADL scores stabilized at a plateau for about 6 months and then continued to decline. Moreover, at 12 months, 58% of the patients had died.
The MDS-ADL score is based on seven components: eating, bed mobility, locomotion, transferring, toileting, hygiene, and dressing; function declined in all of these areas when patients started dialysis.
Patients were more likely to decline in activities of daily living after starting dialysis if they were older, were white, had cerebrovascular disease, had a diagnosis of dementia, were hospitalized at the start of dialysis, or had a serum albumin level lower than 3.5 g/dL.
The same thing happens to elders living in the community when they start dialysis. Jassal and colleagues12 reported that, of 97 community-dwelling patients (mean age 85), 46 (47%) were dead 2 years after starting dialysis. Although 76 (78%) had been living independently at the start of dialysis, only 11 (11%) were still doing so at 2 years.
Comment. These findings indicate that we do not know if hemodialysis improves survival. Hemodialysis may buy about 3 months of stable function, but it clearly does not restore function.
Is this the best we can do? Standard hemodialysis may have flaws, and nocturnal dialysis and peritoneal dialysis are used more in other countries. These dialysis techniques require more study in our older population. The lesson from these two publications on dialysis is that we should attend more carefully to slowing the decline in renal function before patients reach end-stage renal disease.
DABIGATRAN: AN ALTERNATIVE TO WARFARIN FOR ATRIAL FIBRILLATION
CONNOLLY SJ, EZEKOWITZ MD, YUSUF S, ET AL; RE-LY STEERING COMMITTEE AND INVESTIGATORS. DABIGATRAN VERSUS WARFARIN IN PATIENTS WITH ATRIAL FIBRILLATION. N ENGL J MED 2009; 361:1139–1151.
Atrial fibrillation is common, affecting 2.2 million adults. The median age of people who have atrial fibrillation is 75 years, and it is the most common arrhythmia in the elderly. Some 20% of ischemic strokes are attributed to it.13–15
Warfarin (Coumadin) is still the mainstay of treatment to prevent stroke in patients with atrial fibrillation. In an analysis of pooled data from five clinical trials,16 the relative risk reduction with warfarin was about 68% in the overall population (number needed to treat 32), 51% in people older than 75 years with no other risk factors (number needed to treat 56), and 85% in people older than 75 years with one or more risk factors (number needed to treat 15).
But warfarin carries a risk of bleeding, and its dose must be periodically adjusted on the basis of the international normalized ratio (INR) of the prothrombin time, so it carries a burden of laboratory monitoring. It is less safe in people who eat erratically, resulting in wide fluctuations in the INR.
Dabigatran (Pradaxa), a direct thrombin inhibitor, is expected to become an alternative to warfarin. It has been approved in Europe but not yet in the United States.
Connolly et al,17 in a randomized, double-blind trial, assigned 18,113 patients who had atrial fibrillation to receive either dabigatran 110 or 150 mg twice daily or adjusted-dose warfarin in an unblinded fashion. At 2 years, the rates of stroke and systemic embolism were about the same with dabigatran 110 mg as with warfarin but were lower with dabigatran 150 mg (relative risk 0.66, 95% confidence interval [CI] 0.53–0.82, P < .001). The rate of major bleeding was lower with dabigatran 110 mg than with warfarin (2.71% per year vs 3.36% per year, P = .003), but it was similar with dabigatran 150 mg (3.11% per year). Rates of life-threatening bleeding were 1.80% with warfarin, 1.22% with dabigatran 110 mg (P < .05), and 1.45% with dabigatran 150 mg (P < .05).
Comment. I suspect that warfarin’s days are numbered. Dabigatran 110 or 150 mg was as safe and as effective as warfarin in clinical trials, and probably will be more effective than warfarin in clinical practice. It will also probably be safer than warfarin in clinical practice, particularly in challenging settings such as long-term care. On the other hand, it will likely be much more expensive than warfarin.
DEMENTIA
Adverse effects of cholinesterase inhibitors
GILL SS, ANDERSON GM, FISCHER HD, ET AL. SYNCOPE AND ITS CONSEQUENCES IN PATIENTS WITH DEMENTIA RECEIVING CHOLINESTERASE INHIBITORS: A POPULATION-BASED COHORT STUDY. ARCH INTERN MED 2009; 169:867–873.
Cholinesterase inhibitors, eg, donepezil (Aricept), galantamine (Razadyne), and rivastigmine (Exelon), are commonly used to treat Alzheimer disease. However, these drugs carry risks of serious adverse effects.
Gill et al18 retrospectively reviewed a database from Ontario, Canada, and identified about 20,000 community-dwelling elderly persons admitted to the hospital who had been prescribed cholinesterase inhibitors and about three times as many matched controls.
Several adverse events were more frequent in people receiving cholinesterase inhibitors. Findings (events per 1,000 person-years):
Hospital visits for syncope: 31.5 vs 18.6, adjusted hazard ratio (HR) 1.76, 95% CI 1.57–1.98
Hip fractures: 22.4 vs 19.8, HR 1.18, 85% CI 1.04–1.34
Hospital visits for bradycardia: 6.9 vs 4.4, HR 1.69, 95% CI 1.32–2.15
Permanent pacemaker insertion: 4.7 vs 3.3, HR 1.49, 95% CI 1.12–2.00.
Comment. This study adds to the concerns that cholinesterase inhibitors, which have only modest cognitive benefits, may increase the risk of falls, injury, and need for pacemaker placement in demented patients. A low threshold to stop medications in this class should be considered when a patient on a cholinesterase inhibitor presents with bradycardia, falls, and syncope.
The importance of ‘staging’ dementia
IVERSON DJ, GRONSETH GS, REGER MA, ET AL; STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. PRACTICE PARAMETER UPDATE: EVALUATION AND MANAGEMENT OF DRIVING RISK IN DEMENTIA: REPORT OF THE QUALITY STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. NEUROLOGY 2010; 74:1316–1324.
The Clinical Dementia Rating (CDR) is a simple scale that should be applied by clinicians to describe stage of dementia in patients with Alzheimer disease. This scale can be useful in a variety of settings, from prescribing antidementia drugs to determining whether a patient should still drive. Although research protocols utilize a survey or semistructured interview to derive the stage, the clinician can estimate the stage easily in the office, particularly if there is an informant who can comment on performance outside the office.
There are four stages to the CDR19:
0: No dementia
0.5: Mild memory deficit but intact function
1.0: Moderate memory loss with mild functional impairment
2.0: Severe memory loss, moderate functional impairment
3.0: Severe memory loss, no significant function outside of the house.
Comment. The first stage (0.5, mild memory deficit but intact function) corresponds to “mild cognitive impairment.” In the clinic, these patients tend to take more notes. They come to the appointment with a little book and they write everything down so they don’t forget. They do arrive at their appointments on time; they are not crashing the car; they are paying their bills.
Patients with CDR stage 1.0 dementia (moderate memory loss with mild functional impairment) may miss appointments, they may confuse their medications, and they may have problems driving. They are still taking care of their basic needs, and they show up for appointments acceptably washed and dressed. However, they are likely having trouble shopping and managing their finances.
Patients with severe memory loss and moderate functional impairment (CDR stage 2.0) may not realize they haven’t bathed for a week or have worn the same clothes repeatedly. They are having trouble with basic activities of daily living, such as bathing and toilet hygiene. However, if you were to encounter them socially and didn’t talk to them for too long, you might think they were normal.
Those with severe memory loss and no significant function outside the house (CDR stage 3.0) are the most severely disabled. Dementia in these individuals is recognizable at a glance, from across the room.
Alzheimer patients progress through the stages, from CDR stage 0.5 at about 1 year to stage 1 by about 2 years, to stage 2 by 5 years, and to stage 3 at 8 or 9 years.20
In prescribing antidementia medications. The CDR can help with prescribing antidementia drugs. No medications are approved by the FDA for stage 0 or 0.5. Cholinesterase inhibitors are approved for stages 1, 2, and 3; memantine (Namenda) is approved for stages 2 and 3.
Advising about driving. The CDR is the only risk predictor with a quality-of-evidence rating of A. More than half of people with stage 0.5 memory impairment are safe drivers; fewer than half of those with stage 1.0 are still safe drivers; and patients with stage 2.0 dementia should not be driving at all.21 An adverse rating by a caregiver carries a quality-of-evidence rating of B. Predictors of driving risk with a quality-of-evidence rating of C are decreased mileage due to self-restriction, agitation, or aggression; a crash in the past 1 to 5 years; a citation in the past 2 to 3 years; and a Folstein Mini-Mental State Examination score of 24 or less. Studies also show that a memory-impaired person’s self-rating of safe driving ability or of assurance that he or she avoids unsafe situations is not reliable.21
DELIRIUM
Delirium goes by a number of synonyms, eg, “sundowning,” acute confusional state, acute change in mental status, metabolic encephalopathy, toxic encephalopathy (psychosis), acute brain syndrome, and acute toxic psychosis.
Delirium is common in hospitalized elderly patients, occurring in 11% to 42% of elderly hospitalized patients overall, up to 53% of elderly surgical patients on regular hospital floors, 80% of elderly surgical patients in intensive care, and about half of elderly patients after undergoing coronary artery bypass grafting. Unfortunately, it is undiagnosed in 30% to 60% of cases.22–24
Many pathways can lead to delirium, including hypoxemia, metabolic derangement, drug effects, systemic inflammation, and infection.25
Outcomes can vary from full recovery to death. After 1 year, 50% of those who leave the hospital with some evidence of delirium have not regained their baseline function. Delirium also increases the cost of care and the risk of institutionalization.
Delirium can accelerate dementia
FONG TG, JONES RN, SHI P, ET AL. DELIRIUM ACCELERATES COGNITIVE DECLINE IN ALZHEIMER DISEASE. NEUROLOGY 2009; 72:1570–1575.
Delirium accelerates the course of dementia in patients who had some evidence of dementia before they entered the hospital. Often, the change is noticeable by the family.26
Preventing delirium
INOUYE SK BOGARDUS ST JR, CHARPENTIER PA, ET AL. A MULTICOMPONENT INTERVENTION TO PREVENT DELIRIUM IN HOSPITALIZED OLDER PATIENTS. N ENGL J MED 1999; 340:669–676.
LUNDSTRÖM M, OLOFSSON B, STENVALL M, ET AL. POSTOPERATIVE DELIRIUM IN OLD PATIENTS WITH FEMORAL NECK FRACTURE: A RANDOMIZED INTERVENTION STUDY. AGING CLIN EXP RES 2007; 19:178–186.
Delirium can often be prevented. In a report published in 1999, Inouye et al27 described the outcomes of a program to prevent delirium in hospitalized medically ill elderly patients. Interventions were aimed at optimizing cognitive function, preventing sleep deprivation, avoiding immobility, improving vision and hearing, and treating dehydration. The incidence of delirium was 9.9% in the intervention group vs 15% in the control group, a 40% reduction (P < .05).
Lundström et al28 implemented a similar program for elderly patients with hip fractures. Interventions included staff education and teamwork; active prevention, detection, and treatment of delirium; transfusions if hemoglobin levels were less than 10 g/dL; prompt removal of indwelling urinary catheters, with screening for urinary retention; active prevention and treatment of constipation; and protein-enriched meals. The incidence of delirium was 55% in the intervention group vs 75% in the control group, a 27% reduction.
Comment. Although we have long known that the risk of delirium in medical and surgical patients can be reduced, most hospitals do not have systematic programs to detect delirium and reduce its incidence. Hopefully, reduction in delirium risk will also reduce its adverse consequences, including worsening of dementia and increased mortality.
New clinical trials and observational studies are shedding light on ways to improve the health of elderly patients. Here is a brief summary of these trials and how they might influence your clinical practice.
EXERCISE HAS NEWLY DISCOVERED BENEFITS
According to government data,1 exercise has a dose-dependent effect on rates of all-cause mortality: the more hours one exercises per week, the lower the risk of death. The difference in risk is most pronounced as one goes from no exercise to about 3 hours of exercise per week; above 3 hours per week, the curve flattens out but continues to decline. Hence, we advise patients to engage in about 30 minutes of moderate-intensity exercise every day.
Lately, physical exercise has been found to have other, unexpected benefits.
Exercise helps cognition
ERICKSON KI, PRAKASH RS, VOSS MW, ET AL. AEROBIC FITNESS IS ASSOCIATED WITH HIPPOCAMPAL VOLUME IN ELDERLY HUMANS. HIPPOCAMPUS 2009; 19:1030–1039.
ETGEN T, SANDER D, HUNTGEBURTH U, POPPERT H, FÖRSTL H, BICKEL H. PHYSICAL ACTIVITY AND INCIDENT COGNITIVE IMPAIRMENT IN ELDERLY PERSONS: THE INVADE STUDY. ARCH INTERN MED 2010; 170:186–193.
The hippocampus is a structure deep in the brain that is involved in short-term memory. It atrophies with age, more so with dementia. Erickson2 found a correlation between aerobic fitness (as measured by maximum oxygen consumption), hippocampal volume, and spatial memory performance.
Etgen and colleagues3 studied nearly 4,000 older adults in Bavaria for 2 years. Among those reporting no physical activity, 21.4% had cognitive impairment at baseline, compared with 7.3% of those with high activity at baseline. Following those without cognitive impairment over a 2-year period, they found the incidence of new cognitive impairment was 13.9% in those with no physical activity at baseline, 6.7% in those with moderate activity, and 5.1% in those with high activity.
Exercise boosts the effect of influenza vaccine
WOODS JA, KEYLOCK KT, LOWDER T, ET AL. CARDIOVASCULAR EXERCISE TRAINING EXTENDS INFLUENZA VACCINE SEROPROTECTION IN SEDENTARY OLDER ADULTS: THE IMMUNE FUNCTION INTERVENTION TRIAL. J AM GERIATR SOC 2009; 57:2183–2191.
In a study in 144 sedentary but healthy older adults (ages 60 to 83), Woods et al4 randomized the participants to undergo either flexibility or cardiovascular training for 10 months, starting 4 months before their annual influenza shot. Exercise extended the duration of antibody protection, with more participants in the cardiovascular group than in the flexibility group showing protection at 24 weeks against all three strains covered by the vaccine: H1N1, H3N2, and influenza B.
PREVENTING FRACTURES
Each year, about 30% of people age 65 or older fall, sustaining serious injuries in 5% to 10% of cases. Unintentional falls are the main cause of hip fractures, which number 300,000 per year. They are also a common cause of death.
Vitamin D prevents fractures, but can there be too much of a good thing?
BISCHOFF-FERRARI HA, WILLETT WC, WONG JB, ET AL. PREVENTION OF NONVERTEBRAL FRACTURES WITH ORAL VITAMIN D AND DOSE DEPENDENCY: A META-ANALYSIS OF RANDOMIZED CONTROLLED TRIALS. ARCH INTERN MED 2009; 169:551–561.
SANDERS KM, STUART AL, WILLIAMSON EJ, ET AL. ANNUAL HIGH-DOSE ORAL VITAMIN D AND FALLS AND FRACTURES IN OLDER WOMEN: A RANDOMIZED CONTROLLED TRIAL. JAMA 2010; 303:1815–1822.
Bischoff-Ferrari5 performed a meta-analysis of 12 randomized controlled trials of oral supplemental vitamin D3 for preventing nonvertebral fractures in people age 65 and older, and eight trials for preventing hip fractures in the same age group. They found that the higher the daily dose of vitamin D, the lower the relative risk of hip fracture. The threshold dose at which supplementation significantly reduced the risk of falling was about 400 units per day. Higher doses of vitamin D reduced both falls and hip fractures by about 20%. The maximal effect was seen with studies using the maximum daily doses, ie, 770 to 800 units per day—not megadoses, but more than most Americans are taking. The threshold serum level of vitamin D of significance was 60 nmol/L (24 ng/mL).
Of interest, the effect on fractures was independent of calcium supplementation. This is important because calcium supplementation over and above ordinary dietary intake may increase the risk of cardiovascular events.6,7
Despite the benefits of vitamin D, too much may be too much of a good thing. Sanders et al8 performed a double-blind, placebo-controlled trial in 2,256 community-dwelling women, age 70 or older, who were considered to be at high risk for fractures. Half received a large oral dose (500,000 units) once a year for 3 to 5 years, and half got placebo. Their initial serum vitamin D level was 49 nmol/L; the level 30 days after a dose in the treatment group was 120 nmol/L.
Contrary to expectations, the incidence of falls was 15% higher in the vitamin D group than in the placebo group (P = .03), and the incidence of fractures was 26% higher (P = .047). The falls and fractures tended to cluster in the first 3 months after the dose in the active treatment group, when serum vitamin D levels were highest.
Comments. Unless future studies suggest a benefit to megadoses of vitamin D or prove calcium supplementation greater than 1,000 mg is safe, the optimal daily intake of vitamin D is likely 1,000 units, with approximately 200 units from diet and 800 units from supplements. A diet rich in low-fat dairy products may not require calcium supplementation. In those consuming a low-calcium diet, supplements of 500 to 1,000 mg/day are likely adequate.
Denosumab, a new drug for preventing fractures
CUMMINGS SR, SAN MARTIN J, MCCLUNG MR, ET AL; FREEDOM TRIAL. DENOSUMAB FOR PREVENTION OF FRACTURES IN POSTMENOPAUSAL WOMEN WITH OSTEOPOROSIS. N ENGL J MED 2009; 361:756–765.
SMITH MR, EGERDIE B, HERNÁNDEZ TORIZ N, ET AL; DENOSUMAB HALT PROSTATE CANCER STUDY GROUP. DENOSUMAB IN MEN RECEIVING ANDROGEN-DEPRIVATION THERAPY FOR PROSTATE CANCER. N ENGL J MED 2009; 361:745–755.
Denosumab (Prolia) is the first of a new class of drugs for the treatment of osteoporosis. It is a monoclonal antibody and member of the tumor necrosis factor superfamily that binds to the receptor activator nuclear factor kappa B (RANK) ligand. It has an antiresorptive effect, preventing osteoclast differentiation and activation. It is given by subcutaneous injection of 60 mg every 6 months; it is cleared by a nonrenal mechanism.
In a randomized controlled trial in 7,868 women between the ages of 60 and 90 who had osteoporosis, Cummings et al9 reported that denosumab reduced the 3-year incidence of vertebral fractures by 68% (P < .001), reduced the incidence of hip fractures by 40% (P = .01), and reduced the incidence of nonvertebral fractures by 20% (P = .01). In a trial in men receiving androgen deprivation therapy for prostate cancer, Smith et al10 reported that denosumab reduced the incidence of vertebral fracture by 62% (P = .006).
Comment. Denosumab was approved by the US Food and Drug Administration (FDA) on June 1, 2010, and is emerging in specialty clinics at the time of this publication. Its potential impact on clinical care is not yet known. It is costly—about $825 (average wholesale price) per injection—but since it is given by injection it may be easier than a yearly infusion of zoledronic acid (Reclast). It has the potential to suppress immune function, although this was not reported in the clinical trials. It may ultimately have a role in treating osteoporosis in men and women, prostate cancer following androgen deprivation, metastatic prostate cancer, metastatic breast cancer, osteoporosis with renal impairment, and other diseases.
DIALYSIS IN THE ELDERLY: A BLEAK STORY
KURELLA TAMURA M, COVINSKY KE, CHERTOW GM, YAFFE K, LANDEFELD CS, MCCOLLOCH CE. FUNCTIONAL STATUS OF ELDERLY ADULTS BEFORE AND AFTER INITIATION OF DIALYSIS. N ENGL J MED 2009; 361:1539–1547.
JASSAL SV, CHIU E, HLADUNEWITH M. LOSS OF INDEPENDENCE IN PATIENTS STARTING DIALYSIS AT 80 YEARS OF AGE OR OLDER (LETTER). N ENGL J MED 2009; 361:1612–1613.
Nursing home residents account for 4% of all patients in end-stage renal disease. However, the benefits of dialysis in older patients are uncertain. The mortality rate during the first year of dialysis is 35% in patients 70 years of age and older and 50% in patients 80 years and older.
Is dialysis helpful in the elderly, ie, does it improve survival and function?
Kurella Tamura et al11 retrospectively identified 3,702 nursing home residents starting dialysis in whom functional assessments had been done. The numbers told a bleak story. Initiation of dialysis was associated with a sharp decline in functional status, as reflected in an increase of 2.8 points on the 28-point Minimum Data Set–Activities of Daily Living (MDS-ADL) scale (the higher the score, the worse the function). MDS-ADL scores stabilized at a plateau for about 6 months and then continued to decline. Moreover, at 12 months, 58% of the patients had died.
The MDS-ADL score is based on seven components: eating, bed mobility, locomotion, transferring, toileting, hygiene, and dressing; function declined in all of these areas when patients started dialysis.
Patients were more likely to decline in activities of daily living after starting dialysis if they were older, were white, had cerebrovascular disease, had a diagnosis of dementia, were hospitalized at the start of dialysis, or had a serum albumin level lower than 3.5 g/dL.
The same thing happens to elders living in the community when they start dialysis. Jassal and colleagues12 reported that, of 97 community-dwelling patients (mean age 85), 46 (47%) were dead 2 years after starting dialysis. Although 76 (78%) had been living independently at the start of dialysis, only 11 (11%) were still doing so at 2 years.
Comment. These findings indicate that we do not know if hemodialysis improves survival. Hemodialysis may buy about 3 months of stable function, but it clearly does not restore function.
Is this the best we can do? Standard hemodialysis may have flaws, and nocturnal dialysis and peritoneal dialysis are used more in other countries. These dialysis techniques require more study in our older population. The lesson from these two publications on dialysis is that we should attend more carefully to slowing the decline in renal function before patients reach end-stage renal disease.
DABIGATRAN: AN ALTERNATIVE TO WARFARIN FOR ATRIAL FIBRILLATION
CONNOLLY SJ, EZEKOWITZ MD, YUSUF S, ET AL; RE-LY STEERING COMMITTEE AND INVESTIGATORS. DABIGATRAN VERSUS WARFARIN IN PATIENTS WITH ATRIAL FIBRILLATION. N ENGL J MED 2009; 361:1139–1151.
Atrial fibrillation is common, affecting 2.2 million adults. The median age of people who have atrial fibrillation is 75 years, and it is the most common arrhythmia in the elderly. Some 20% of ischemic strokes are attributed to it.13–15
Warfarin (Coumadin) is still the mainstay of treatment to prevent stroke in patients with atrial fibrillation. In an analysis of pooled data from five clinical trials,16 the relative risk reduction with warfarin was about 68% in the overall population (number needed to treat 32), 51% in people older than 75 years with no other risk factors (number needed to treat 56), and 85% in people older than 75 years with one or more risk factors (number needed to treat 15).
But warfarin carries a risk of bleeding, and its dose must be periodically adjusted on the basis of the international normalized ratio (INR) of the prothrombin time, so it carries a burden of laboratory monitoring. It is less safe in people who eat erratically, resulting in wide fluctuations in the INR.
Dabigatran (Pradaxa), a direct thrombin inhibitor, is expected to become an alternative to warfarin. It has been approved in Europe but not yet in the United States.
Connolly et al,17 in a randomized, double-blind trial, assigned 18,113 patients who had atrial fibrillation to receive either dabigatran 110 or 150 mg twice daily or adjusted-dose warfarin in an unblinded fashion. At 2 years, the rates of stroke and systemic embolism were about the same with dabigatran 110 mg as with warfarin but were lower with dabigatran 150 mg (relative risk 0.66, 95% confidence interval [CI] 0.53–0.82, P < .001). The rate of major bleeding was lower with dabigatran 110 mg than with warfarin (2.71% per year vs 3.36% per year, P = .003), but it was similar with dabigatran 150 mg (3.11% per year). Rates of life-threatening bleeding were 1.80% with warfarin, 1.22% with dabigatran 110 mg (P < .05), and 1.45% with dabigatran 150 mg (P < .05).
Comment. I suspect that warfarin’s days are numbered. Dabigatran 110 or 150 mg was as safe and as effective as warfarin in clinical trials, and probably will be more effective than warfarin in clinical practice. It will also probably be safer than warfarin in clinical practice, particularly in challenging settings such as long-term care. On the other hand, it will likely be much more expensive than warfarin.
DEMENTIA
Adverse effects of cholinesterase inhibitors
GILL SS, ANDERSON GM, FISCHER HD, ET AL. SYNCOPE AND ITS CONSEQUENCES IN PATIENTS WITH DEMENTIA RECEIVING CHOLINESTERASE INHIBITORS: A POPULATION-BASED COHORT STUDY. ARCH INTERN MED 2009; 169:867–873.
Cholinesterase inhibitors, eg, donepezil (Aricept), galantamine (Razadyne), and rivastigmine (Exelon), are commonly used to treat Alzheimer disease. However, these drugs carry risks of serious adverse effects.
Gill et al18 retrospectively reviewed a database from Ontario, Canada, and identified about 20,000 community-dwelling elderly persons admitted to the hospital who had been prescribed cholinesterase inhibitors and about three times as many matched controls.
Several adverse events were more frequent in people receiving cholinesterase inhibitors. Findings (events per 1,000 person-years):
Hospital visits for syncope: 31.5 vs 18.6, adjusted hazard ratio (HR) 1.76, 95% CI 1.57–1.98
Hip fractures: 22.4 vs 19.8, HR 1.18, 85% CI 1.04–1.34
Hospital visits for bradycardia: 6.9 vs 4.4, HR 1.69, 95% CI 1.32–2.15
Permanent pacemaker insertion: 4.7 vs 3.3, HR 1.49, 95% CI 1.12–2.00.
Comment. This study adds to the concerns that cholinesterase inhibitors, which have only modest cognitive benefits, may increase the risk of falls, injury, and need for pacemaker placement in demented patients. A low threshold to stop medications in this class should be considered when a patient on a cholinesterase inhibitor presents with bradycardia, falls, and syncope.
The importance of ‘staging’ dementia
IVERSON DJ, GRONSETH GS, REGER MA, ET AL; STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. PRACTICE PARAMETER UPDATE: EVALUATION AND MANAGEMENT OF DRIVING RISK IN DEMENTIA: REPORT OF THE QUALITY STANDARDS SUBCOMMITTEE OF THE AMERICAN ACADEMY OF NEUROLOGY. NEUROLOGY 2010; 74:1316–1324.
The Clinical Dementia Rating (CDR) is a simple scale that should be applied by clinicians to describe stage of dementia in patients with Alzheimer disease. This scale can be useful in a variety of settings, from prescribing antidementia drugs to determining whether a patient should still drive. Although research protocols utilize a survey or semistructured interview to derive the stage, the clinician can estimate the stage easily in the office, particularly if there is an informant who can comment on performance outside the office.
There are four stages to the CDR19:
0: No dementia
0.5: Mild memory deficit but intact function
1.0: Moderate memory loss with mild functional impairment
2.0: Severe memory loss, moderate functional impairment
3.0: Severe memory loss, no significant function outside of the house.
Comment. The first stage (0.5, mild memory deficit but intact function) corresponds to “mild cognitive impairment.” In the clinic, these patients tend to take more notes. They come to the appointment with a little book and they write everything down so they don’t forget. They do arrive at their appointments on time; they are not crashing the car; they are paying their bills.
Patients with CDR stage 1.0 dementia (moderate memory loss with mild functional impairment) may miss appointments, they may confuse their medications, and they may have problems driving. They are still taking care of their basic needs, and they show up for appointments acceptably washed and dressed. However, they are likely having trouble shopping and managing their finances.
Patients with severe memory loss and moderate functional impairment (CDR stage 2.0) may not realize they haven’t bathed for a week or have worn the same clothes repeatedly. They are having trouble with basic activities of daily living, such as bathing and toilet hygiene. However, if you were to encounter them socially and didn’t talk to them for too long, you might think they were normal.
Those with severe memory loss and no significant function outside the house (CDR stage 3.0) are the most severely disabled. Dementia in these individuals is recognizable at a glance, from across the room.
Alzheimer patients progress through the stages, from CDR stage 0.5 at about 1 year to stage 1 by about 2 years, to stage 2 by 5 years, and to stage 3 at 8 or 9 years.20
In prescribing antidementia medications. The CDR can help with prescribing antidementia drugs. No medications are approved by the FDA for stage 0 or 0.5. Cholinesterase inhibitors are approved for stages 1, 2, and 3; memantine (Namenda) is approved for stages 2 and 3.
Advising about driving. The CDR is the only risk predictor with a quality-of-evidence rating of A. More than half of people with stage 0.5 memory impairment are safe drivers; fewer than half of those with stage 1.0 are still safe drivers; and patients with stage 2.0 dementia should not be driving at all.21 An adverse rating by a caregiver carries a quality-of-evidence rating of B. Predictors of driving risk with a quality-of-evidence rating of C are decreased mileage due to self-restriction, agitation, or aggression; a crash in the past 1 to 5 years; a citation in the past 2 to 3 years; and a Folstein Mini-Mental State Examination score of 24 or less. Studies also show that a memory-impaired person’s self-rating of safe driving ability or of assurance that he or she avoids unsafe situations is not reliable.21
DELIRIUM
Delirium goes by a number of synonyms, eg, “sundowning,” acute confusional state, acute change in mental status, metabolic encephalopathy, toxic encephalopathy (psychosis), acute brain syndrome, and acute toxic psychosis.
Delirium is common in hospitalized elderly patients, occurring in 11% to 42% of elderly hospitalized patients overall, up to 53% of elderly surgical patients on regular hospital floors, 80% of elderly surgical patients in intensive care, and about half of elderly patients after undergoing coronary artery bypass grafting. Unfortunately, it is undiagnosed in 30% to 60% of cases.22–24
Many pathways can lead to delirium, including hypoxemia, metabolic derangement, drug effects, systemic inflammation, and infection.25
Outcomes can vary from full recovery to death. After 1 year, 50% of those who leave the hospital with some evidence of delirium have not regained their baseline function. Delirium also increases the cost of care and the risk of institutionalization.
Delirium can accelerate dementia
FONG TG, JONES RN, SHI P, ET AL. DELIRIUM ACCELERATES COGNITIVE DECLINE IN ALZHEIMER DISEASE. NEUROLOGY 2009; 72:1570–1575.
Delirium accelerates the course of dementia in patients who had some evidence of dementia before they entered the hospital. Often, the change is noticeable by the family.26
Preventing delirium
INOUYE SK BOGARDUS ST JR, CHARPENTIER PA, ET AL. A MULTICOMPONENT INTERVENTION TO PREVENT DELIRIUM IN HOSPITALIZED OLDER PATIENTS. N ENGL J MED 1999; 340:669–676.
LUNDSTRÖM M, OLOFSSON B, STENVALL M, ET AL. POSTOPERATIVE DELIRIUM IN OLD PATIENTS WITH FEMORAL NECK FRACTURE: A RANDOMIZED INTERVENTION STUDY. AGING CLIN EXP RES 2007; 19:178–186.
Delirium can often be prevented. In a report published in 1999, Inouye et al27 described the outcomes of a program to prevent delirium in hospitalized medically ill elderly patients. Interventions were aimed at optimizing cognitive function, preventing sleep deprivation, avoiding immobility, improving vision and hearing, and treating dehydration. The incidence of delirium was 9.9% in the intervention group vs 15% in the control group, a 40% reduction (P < .05).
Lundström et al28 implemented a similar program for elderly patients with hip fractures. Interventions included staff education and teamwork; active prevention, detection, and treatment of delirium; transfusions if hemoglobin levels were less than 10 g/dL; prompt removal of indwelling urinary catheters, with screening for urinary retention; active prevention and treatment of constipation; and protein-enriched meals. The incidence of delirium was 55% in the intervention group vs 75% in the control group, a 27% reduction.
Comment. Although we have long known that the risk of delirium in medical and surgical patients can be reduced, most hospitals do not have systematic programs to detect delirium and reduce its incidence. Hopefully, reduction in delirium risk will also reduce its adverse consequences, including worsening of dementia and increased mortality.
Erickson KI, Prakash RS, Voss MW, et al. Aerobic fitness is associated with hippocampal volume in elderly humans. Hippocampus2009; 19:1030–1039.
Etgen T, Sander D, Huntgeburth U, Poppert H, Förstl H, Bickel H. Physical activity and incident cognitive impairment in elderly persons: the INVADE study. Arch Intern Med2010; 170:186–193.
Woods JA, Keylock KT, Lowder T, et al. Cardiovascular exercise training extends influenza vaccine seroprotection in sedentary older adults: the immune function intervention trial. J Am Geriatr Soc2009; 57:2183–2191.
Bischoff-Ferrari HA, Willett WC, Wong JB, et al. Prevention of nonvertebral fractures with oral vitamin D and dose dependency: a meta-analysis of randomized controlled trials. Arch Intern Med2009; 169:551–561.
Bolland MJ, Avenell A, Baron JA, et al. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. BMJ2010; 341:c3691. doi:10.1136/bmj.c3691.
Bolland MJ, Barber PA, Doughty RN, et al. Vascular events in healthy older women receiving calcium supplementation: randomised controlled trial. BMJ2008; 336:262–266.
Sanders KM, Stuart AL, Williamson EJ, et al. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA2010; 303:1815–1822.
Cummings SR, San Martin J, McClung MR, et al; FREEDOM Trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med2009; 361:756–765.
Smith MR, Egerdie B, Hernández Toriz N, et al; Denosumab HALT Prostate Cancer Study Group. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med2009; 361:745–755.
Kurella Tamura M, Covinsky KE, Chertow GM, Yaffe K, Landefeld CS, McColloch CE. Functional status of elderly adults before and after initiation of dialysis. N Engl J Med2009; 361:1539–1547.
Jassal SV, Chiu E, Hladunewich M. Loss of independence in patients starting dialysis at 80 years of age or older (letter). N Engl J Med2009; 361:1612–1613.
Feinberg WM, Blackshear JL, Laupacis A, Kronmal R, Hart RG. Prevalence, age distribution and gender of patients with atrial fibrillation. Analysis and implications. Arch Intern Med1995; 155:469–473.
Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation: a major contributor to stroke in the elderly. The Framingham Study. Arch Intern Med1987; 147:1561–1564.
Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial fibrillation. The Framingham Study. Stroke1996; 27:1760–1764.
Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med1994; 154:1449–1457.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern Med2009; 169:867–873.
Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology1993; 43:2412–2414.
Sloane PD. Advances in the treatment of Alzheimer’s disease. Am Fam Physician1998; 58:1577–1586.
Iverson DJ, Gronseth GS, Reger MA, et al; Standards Subcommittee of the American Academy of Neurology. Practice parameter update: evaluation and management of driving risk in dementia: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology2010; 74:1316–1324.
Demeure MJ, Fain MJ. The elderly surgical patient and postoperative delirium. J Am Coll Surg2006; 203:752–757.
Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing2006; 35:350–364.
Rudolph JL, Jones RN, Levkoff SE, et al. Derivation and validation of a preoperative prediction rule for delirium after cardiac surgery. Circulation2009; 119:229–236.
Fong TG, Tulebaev SR, Inouye SK. Delirium in elderly adults: diagnosis, prevention and treatment. Nat Rev Neurol2009; 5:210–220.
Fong TG, Jones RN, Shi P, et al. Delirium accelerates cognitive decline in Alzheimer disease. Neurology2009; 72:1570–1575.
Inouye SK, Bogardus ST, Charpentier PA, et al. A multicomponent intervention to prevent delirium in hospitalized older patients. N Engl J Med1999; 340:669–676.
Lundström M, Olofsson B, Stenvall M, et al. Postoperative delirium in old patients with femoral neck fracture: a randomized intervention study. Aging Clin Exp Res2007; 19:178–186.
Erickson KI, Prakash RS, Voss MW, et al. Aerobic fitness is associated with hippocampal volume in elderly humans. Hippocampus2009; 19:1030–1039.
Etgen T, Sander D, Huntgeburth U, Poppert H, Förstl H, Bickel H. Physical activity and incident cognitive impairment in elderly persons: the INVADE study. Arch Intern Med2010; 170:186–193.
Woods JA, Keylock KT, Lowder T, et al. Cardiovascular exercise training extends influenza vaccine seroprotection in sedentary older adults: the immune function intervention trial. J Am Geriatr Soc2009; 57:2183–2191.
Bischoff-Ferrari HA, Willett WC, Wong JB, et al. Prevention of nonvertebral fractures with oral vitamin D and dose dependency: a meta-analysis of randomized controlled trials. Arch Intern Med2009; 169:551–561.
Bolland MJ, Avenell A, Baron JA, et al. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. BMJ2010; 341:c3691. doi:10.1136/bmj.c3691.
Bolland MJ, Barber PA, Doughty RN, et al. Vascular events in healthy older women receiving calcium supplementation: randomised controlled trial. BMJ2008; 336:262–266.
Sanders KM, Stuart AL, Williamson EJ, et al. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA2010; 303:1815–1822.
Cummings SR, San Martin J, McClung MR, et al; FREEDOM Trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med2009; 361:756–765.
Smith MR, Egerdie B, Hernández Toriz N, et al; Denosumab HALT Prostate Cancer Study Group. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med2009; 361:745–755.
Kurella Tamura M, Covinsky KE, Chertow GM, Yaffe K, Landefeld CS, McColloch CE. Functional status of elderly adults before and after initiation of dialysis. N Engl J Med2009; 361:1539–1547.
Jassal SV, Chiu E, Hladunewich M. Loss of independence in patients starting dialysis at 80 years of age or older (letter). N Engl J Med2009; 361:1612–1613.
Feinberg WM, Blackshear JL, Laupacis A, Kronmal R, Hart RG. Prevalence, age distribution and gender of patients with atrial fibrillation. Analysis and implications. Arch Intern Med1995; 155:469–473.
Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation: a major contributor to stroke in the elderly. The Framingham Study. Arch Intern Med1987; 147:1561–1564.
Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial fibrillation. The Framingham Study. Stroke1996; 27:1760–1764.
Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med1994; 154:1449–1457.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern Med2009; 169:867–873.
Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology1993; 43:2412–2414.
Sloane PD. Advances in the treatment of Alzheimer’s disease. Am Fam Physician1998; 58:1577–1586.
Iverson DJ, Gronseth GS, Reger MA, et al; Standards Subcommittee of the American Academy of Neurology. Practice parameter update: evaluation and management of driving risk in dementia: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology2010; 74:1316–1324.
Demeure MJ, Fain MJ. The elderly surgical patient and postoperative delirium. J Am Coll Surg2006; 203:752–757.
Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing2006; 35:350–364.
Rudolph JL, Jones RN, Levkoff SE, et al. Derivation and validation of a preoperative prediction rule for delirium after cardiac surgery. Circulation2009; 119:229–236.
Fong TG, Tulebaev SR, Inouye SK. Delirium in elderly adults: diagnosis, prevention and treatment. Nat Rev Neurol2009; 5:210–220.
Fong TG, Jones RN, Shi P, et al. Delirium accelerates cognitive decline in Alzheimer disease. Neurology2009; 72:1570–1575.
Inouye SK, Bogardus ST, Charpentier PA, et al. A multicomponent intervention to prevent delirium in hospitalized older patients. N Engl J Med1999; 340:669–676.
Lundström M, Olofsson B, Stenvall M, et al. Postoperative delirium in old patients with femoral neck fracture: a randomized intervention study. Aging Clin Exp Res2007; 19:178–186.
Exercise has newly discovered benefits, such as preserving cognition and boosting the response to vaccination.
Vitamin D supplementation has been found to prevent fractures, but yearly megadoses had the opposite effect.
Denosumab (Prolia) has been approved for preventing fractures. It acts by inhibiting the receptor activator of nuclear factor kappa B (RANK) ligand.
The outlook for elderly patients starting hemodialysis is bleak, with loss of function and a high risk of death.
Dabigatran (Pradaxa), a direct thrombin inhibitor, may prove to be a safer alternative to warfarin (Coumadin).
Cholinesterase inhibitors for Alzheimer disease are associated with higher risks of hospitalization for syncope, hip fractures, bradycardia, and pacemaker insertion.
The Clinical Dementia Rating should be estimated when prescribing a cognitive enhancer and when advising a patient with memory impairment on driving safety.
Delirium often accelerates dementia; interventions for hospitalized elderly patients may reduce its incidence.
Patients with an osteoporotic hip fracture suffer from profound morbidity and are at a heightened risk of death. It is therefore essential that they receive treatment with a bisphosphonate known to modify the subsequent risk of fracture at any site—eg, alendronate (Fosamax), risedronate (Actonel), or zoledronic acid (Reclast).
However, there is concern that starting a bisphosphonate too soon after surgery could disrupt bone remodeling and delay fracture repair.
Only one clinical study addressed the timing of bisphosphonate therapy after hip fracture repair. In this study, Eriksen et al1 performed a post hoc analysis of data from the Health Outcomes and Reduced Incidence With Zoledronic Acid Once Yearly Recurrent Fracture Trial (HORIZON-RFT)2 and concluded that the optimal time to give intravenous zoledronic acid is 2 to 12 weeks after surgical repair of the fracture.
In a frail, elderly patient with comorbidities, a single intravenous 5-mg dose of zoledronic acid guarantees adequate treatment, obviating issues of poor compliance and oral absorption and loss to follow-up. Sufficient levels of vitamin D and calcium should be ensured.
THE EVIDENCE
The original HORIZON-RFT study,2 published in 2007, compared intravenous zoledronic acid against placebo in elderly patients with osteoporotic hip fracture. Most of the patients were white women; their mean age was 74; 1,065 received intravenous zoledronic acid, and 1,062 received placebo. All received vitamin D and calcium.
The trial showed a clear reduction in the rate of recurrent fractures at other sites (a primary end point) and a reduction in the rate of all-cause mortality in patients treated within 90 days of fracture. A total of 424 fractures occurred in 231 patients. The risk of any new clinical fracture was 35% lower with treatment than with placebo (occurring in 8.6% vs 13.9% of patients, P = .001), and the number of deaths due to any cause was 28% lower with treatment than with placebo (occurring in 101 vs 141, P = .01).2
The mean time to fracture was 39.8 months in the treated group vs 36.4 in the placebo group. The fracture risk reduction began to be apparent by 12 months, and the reduction in mortality rate by 16 months.2
In a post hoc analysis of the trial, Eriksen et al1 attempted to ascertain the optimal time for therapy in terms of fracture risk and mortality reduction. Analyzing the data by 2-week intervals beginning after the surgical repair of the fracture, the authors found that only 56 patients (5.3%) had received zoledronic acid within 2 weeks of surgery and only 47 had received placebo, and they saw no advantage to intravenous zoledronic acid compared with placebo in these first 2 weeks with respect to bone mineral density, fracture risk, or risk of death. However, excluding this small subset, antifracture efficacy and reduction in mortality rate were present when patients were treated with zoledronic acid in the 2 to 12 weeks after hip fracture repair, and improvement in bone mineral density at the hip was noted at 12 months in all cohorts.
Colón-Emeric et al3 performed another post hoc analysis, attempting to explain the lower mortality rate seen in patients treated with zoledronic acid. It had been an unexpected finding, and determinants of mortality rate reduction were hampered by a limited knowledge of the true cause of death or the circumstances of care after fracture. The authors concluded that only 8% of the reduction in mortality rate evident early in the second year of treatment with zoledronic acid could be attributed to a reduction in fractures.3 Other mechanisms by which the mortality rate reduction occurred remained unclear.
Curiously, in another large randomized controlled trial of zoledronic acid, in women with postmenopausal osteoporosis, Black et al4 reported that more patients died in the treated group (130 of 3,862) than in the placebo group (112 of 3,852). This difference was not statistically significant, but neither was it explained.
A meta-analysis by Bolland et al5 examined the effect of other osteoporosis treatments on mortality rate, using randomized controlled trials that lasted more than 12 months and that reported more than 10 deaths. The authors concluded the following:
In the trials in which bisphosphonates reduced the mortality rate, the mortality rate in the placebo group was higher than 10 per 1,000 patient-years
The effect of osteoporosis treatment on the mortality rate in a frail, elderly population is evident using agents with proven efficacy in reducing vertebral and nonvertebral fractures, eg, alendronate, risedronate, and zoledronic acid.5
THE SCIENCE
Osteoporotic fractures occur with minimal trauma, with the failure of bone attributed to impaired integrity of bone microarchitecture. The ultimate goal of fracture repair is to restore bone size, shape, and tissue properties. The issue of when to treat with a bisphosphonate after hip fracture arises because bisphosphonates are known to disrupt bone remodeling and so delay fracture repair.
After fracture, both anabolic and catabolic phases occur.6 The final outcome depends on the following:
The type of intervention to stabilize the fracture site (eg, surgical repair)
The inflammatory cytokines and growth factors released by the cellular elements in bloody and disrupted tissue.
Oxygen tension, angiogenesis, and osteoblasts are critical to primary bone formation, and osteoclasts are essential in remodeling this initial bone deposition. These late phases of fracture repair are most vulnerable to the bisphosphonates, through suppression of osteoclast resorption and possibly through decreased angiogenesis.6 Callus formation is sustained, but bone remodeling is delayed.
Amanat et al7 examined the timing of a single dose of zoledronic acid after fracture repair in a rat model of diaphyseal fracture and found that the callus was larger and stronger if the bisphosphonate dose had been delayed 1 or 2 weeks. The animals treated with zoledronic acid showed a remarkable trabecular network of bone between the original femoral cortex and the new cortical bone that was not present in the control group, perhaps contributing to the enhanced mechanical properties of the callus. Other studies suggest single dosing rather than continuous dosing may be advantageous in fracture healing.8
THE REALITY
Healthy dogs or growing rats with linear diaphyseal fractures are imperfect models for elderly osteoporotic patients with hip fracture, as Dr. Herbert Fleisch noted in his editorial, “Can bisphosphonates be given to patients with fractures?”9 Still, if retained primary bone can be used in the process of fracture repair to gain an early mechanical advantage, then perhaps delayed remodeling will permit early mobilization and further fracture prevention in humans.
How soon after hip fracture surgery should a patient start a bisphosphonate? The only data we have are from a single randomized controlled trial designed to measure fracture risk reduction in osteoporotic patients with hip fracture using intravenous zoledronic acid 5 mg compared with placebo.2 A post hoc analysis of this study1 generated the limited clinical data we have on the optimal timing of the treatment. Linking these study data with the laboratory data, one would intuit that delaying the infusion of zoledronic acid for at least 2 weeks after hip fracture repair would offer a clinical reduction in fracture risk and improvement (or stabilization) in bone mineral density by 12 months, and a reduction in the rate of all-cause mortality beginning at 16 months.
References
Eriksen EF, Lyles KW, Colón-Emeric CS, et al. Antifracture efficacy and reduction of mortality in relation to timing of the first dose of zoledronic acid after hip fracture. J Bone Miner Res2009; 24:1308–1313.
Lyles KW, Colón-Emeric CS, Magaziner JS, et al; for the HORIZON Recurrent Fracture Trial. Zoledronic acid and clinical fractures and mortality after hip fracture. N Engl J Med2007; 357:1799–1809.
Colón-Emeric CS, Mesenbrink P, Lyles KW, et al. Potential mediators of the mortality reduction with zoledronic acid after hip fracture. J Bone Miner Res2010; 25:91–97.
Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med2007; 356:1809–1822.
Bolland MJ, Grey AB, Gamble GD, Reid IR. Effect of osteoporosis treatment on mortality: a meta-analysis. J Clin Endocrinol Metab2010; 95:1174–1181.
Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: the cellular picture. Semin Cell Dev Biol2008; 19:459–466.
Amanat N, McDonald M, Godfrey C, Bilston L, Little D. Optimal timing of a single dose of zoledronic acid to increase strength in rat fracture repair. J Bone Miner Res2007; 22:867–876.
Li J, Mori S, Kaji Y, Mashiba T, Kawanishi J, Norimatsu H. Effect of bisphosphonate (incadronate) on fracture healing of long bones in rats. J Bone Miner Res1999; 14:969–979.
Fleisch H. Can bisphosphonates be given to patients with fractures?J Bone Miner Res2001; 16:437–440.
Margaret Seton, MD Assistant Professor of Medicine, Harvard Medical School; Director, Rheumatology Fellowship Program, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
Address: Margaret Seton, MD, Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Bulfinch 165, 55 Fruit Street, Boston, MA 02114; e-mail [email protected]
Margaret Seton, MD Assistant Professor of Medicine, Harvard Medical School; Director, Rheumatology Fellowship Program, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
Address: Margaret Seton, MD, Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Bulfinch 165, 55 Fruit Street, Boston, MA 02114; e-mail [email protected]
Author and Disclosure Information
Margaret Seton, MD Assistant Professor of Medicine, Harvard Medical School; Director, Rheumatology Fellowship Program, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
Address: Margaret Seton, MD, Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Bulfinch 165, 55 Fruit Street, Boston, MA 02114; e-mail [email protected]
Patients with an osteoporotic hip fracture suffer from profound morbidity and are at a heightened risk of death. It is therefore essential that they receive treatment with a bisphosphonate known to modify the subsequent risk of fracture at any site—eg, alendronate (Fosamax), risedronate (Actonel), or zoledronic acid (Reclast).
However, there is concern that starting a bisphosphonate too soon after surgery could disrupt bone remodeling and delay fracture repair.
Only one clinical study addressed the timing of bisphosphonate therapy after hip fracture repair. In this study, Eriksen et al1 performed a post hoc analysis of data from the Health Outcomes and Reduced Incidence With Zoledronic Acid Once Yearly Recurrent Fracture Trial (HORIZON-RFT)2 and concluded that the optimal time to give intravenous zoledronic acid is 2 to 12 weeks after surgical repair of the fracture.
In a frail, elderly patient with comorbidities, a single intravenous 5-mg dose of zoledronic acid guarantees adequate treatment, obviating issues of poor compliance and oral absorption and loss to follow-up. Sufficient levels of vitamin D and calcium should be ensured.
THE EVIDENCE
The original HORIZON-RFT study,2 published in 2007, compared intravenous zoledronic acid against placebo in elderly patients with osteoporotic hip fracture. Most of the patients were white women; their mean age was 74; 1,065 received intravenous zoledronic acid, and 1,062 received placebo. All received vitamin D and calcium.
The trial showed a clear reduction in the rate of recurrent fractures at other sites (a primary end point) and a reduction in the rate of all-cause mortality in patients treated within 90 days of fracture. A total of 424 fractures occurred in 231 patients. The risk of any new clinical fracture was 35% lower with treatment than with placebo (occurring in 8.6% vs 13.9% of patients, P = .001), and the number of deaths due to any cause was 28% lower with treatment than with placebo (occurring in 101 vs 141, P = .01).2
The mean time to fracture was 39.8 months in the treated group vs 36.4 in the placebo group. The fracture risk reduction began to be apparent by 12 months, and the reduction in mortality rate by 16 months.2
In a post hoc analysis of the trial, Eriksen et al1 attempted to ascertain the optimal time for therapy in terms of fracture risk and mortality reduction. Analyzing the data by 2-week intervals beginning after the surgical repair of the fracture, the authors found that only 56 patients (5.3%) had received zoledronic acid within 2 weeks of surgery and only 47 had received placebo, and they saw no advantage to intravenous zoledronic acid compared with placebo in these first 2 weeks with respect to bone mineral density, fracture risk, or risk of death. However, excluding this small subset, antifracture efficacy and reduction in mortality rate were present when patients were treated with zoledronic acid in the 2 to 12 weeks after hip fracture repair, and improvement in bone mineral density at the hip was noted at 12 months in all cohorts.
Colón-Emeric et al3 performed another post hoc analysis, attempting to explain the lower mortality rate seen in patients treated with zoledronic acid. It had been an unexpected finding, and determinants of mortality rate reduction were hampered by a limited knowledge of the true cause of death or the circumstances of care after fracture. The authors concluded that only 8% of the reduction in mortality rate evident early in the second year of treatment with zoledronic acid could be attributed to a reduction in fractures.3 Other mechanisms by which the mortality rate reduction occurred remained unclear.
Curiously, in another large randomized controlled trial of zoledronic acid, in women with postmenopausal osteoporosis, Black et al4 reported that more patients died in the treated group (130 of 3,862) than in the placebo group (112 of 3,852). This difference was not statistically significant, but neither was it explained.
A meta-analysis by Bolland et al5 examined the effect of other osteoporosis treatments on mortality rate, using randomized controlled trials that lasted more than 12 months and that reported more than 10 deaths. The authors concluded the following:
In the trials in which bisphosphonates reduced the mortality rate, the mortality rate in the placebo group was higher than 10 per 1,000 patient-years
The effect of osteoporosis treatment on the mortality rate in a frail, elderly population is evident using agents with proven efficacy in reducing vertebral and nonvertebral fractures, eg, alendronate, risedronate, and zoledronic acid.5
THE SCIENCE
Osteoporotic fractures occur with minimal trauma, with the failure of bone attributed to impaired integrity of bone microarchitecture. The ultimate goal of fracture repair is to restore bone size, shape, and tissue properties. The issue of when to treat with a bisphosphonate after hip fracture arises because bisphosphonates are known to disrupt bone remodeling and so delay fracture repair.
After fracture, both anabolic and catabolic phases occur.6 The final outcome depends on the following:
The type of intervention to stabilize the fracture site (eg, surgical repair)
The inflammatory cytokines and growth factors released by the cellular elements in bloody and disrupted tissue.
Oxygen tension, angiogenesis, and osteoblasts are critical to primary bone formation, and osteoclasts are essential in remodeling this initial bone deposition. These late phases of fracture repair are most vulnerable to the bisphosphonates, through suppression of osteoclast resorption and possibly through decreased angiogenesis.6 Callus formation is sustained, but bone remodeling is delayed.
Amanat et al7 examined the timing of a single dose of zoledronic acid after fracture repair in a rat model of diaphyseal fracture and found that the callus was larger and stronger if the bisphosphonate dose had been delayed 1 or 2 weeks. The animals treated with zoledronic acid showed a remarkable trabecular network of bone between the original femoral cortex and the new cortical bone that was not present in the control group, perhaps contributing to the enhanced mechanical properties of the callus. Other studies suggest single dosing rather than continuous dosing may be advantageous in fracture healing.8
THE REALITY
Healthy dogs or growing rats with linear diaphyseal fractures are imperfect models for elderly osteoporotic patients with hip fracture, as Dr. Herbert Fleisch noted in his editorial, “Can bisphosphonates be given to patients with fractures?”9 Still, if retained primary bone can be used in the process of fracture repair to gain an early mechanical advantage, then perhaps delayed remodeling will permit early mobilization and further fracture prevention in humans.
How soon after hip fracture surgery should a patient start a bisphosphonate? The only data we have are from a single randomized controlled trial designed to measure fracture risk reduction in osteoporotic patients with hip fracture using intravenous zoledronic acid 5 mg compared with placebo.2 A post hoc analysis of this study1 generated the limited clinical data we have on the optimal timing of the treatment. Linking these study data with the laboratory data, one would intuit that delaying the infusion of zoledronic acid for at least 2 weeks after hip fracture repair would offer a clinical reduction in fracture risk and improvement (or stabilization) in bone mineral density by 12 months, and a reduction in the rate of all-cause mortality beginning at 16 months.
Patients with an osteoporotic hip fracture suffer from profound morbidity and are at a heightened risk of death. It is therefore essential that they receive treatment with a bisphosphonate known to modify the subsequent risk of fracture at any site—eg, alendronate (Fosamax), risedronate (Actonel), or zoledronic acid (Reclast).
However, there is concern that starting a bisphosphonate too soon after surgery could disrupt bone remodeling and delay fracture repair.
Only one clinical study addressed the timing of bisphosphonate therapy after hip fracture repair. In this study, Eriksen et al1 performed a post hoc analysis of data from the Health Outcomes and Reduced Incidence With Zoledronic Acid Once Yearly Recurrent Fracture Trial (HORIZON-RFT)2 and concluded that the optimal time to give intravenous zoledronic acid is 2 to 12 weeks after surgical repair of the fracture.
In a frail, elderly patient with comorbidities, a single intravenous 5-mg dose of zoledronic acid guarantees adequate treatment, obviating issues of poor compliance and oral absorption and loss to follow-up. Sufficient levels of vitamin D and calcium should be ensured.
THE EVIDENCE
The original HORIZON-RFT study,2 published in 2007, compared intravenous zoledronic acid against placebo in elderly patients with osteoporotic hip fracture. Most of the patients were white women; their mean age was 74; 1,065 received intravenous zoledronic acid, and 1,062 received placebo. All received vitamin D and calcium.
The trial showed a clear reduction in the rate of recurrent fractures at other sites (a primary end point) and a reduction in the rate of all-cause mortality in patients treated within 90 days of fracture. A total of 424 fractures occurred in 231 patients. The risk of any new clinical fracture was 35% lower with treatment than with placebo (occurring in 8.6% vs 13.9% of patients, P = .001), and the number of deaths due to any cause was 28% lower with treatment than with placebo (occurring in 101 vs 141, P = .01).2
The mean time to fracture was 39.8 months in the treated group vs 36.4 in the placebo group. The fracture risk reduction began to be apparent by 12 months, and the reduction in mortality rate by 16 months.2
In a post hoc analysis of the trial, Eriksen et al1 attempted to ascertain the optimal time for therapy in terms of fracture risk and mortality reduction. Analyzing the data by 2-week intervals beginning after the surgical repair of the fracture, the authors found that only 56 patients (5.3%) had received zoledronic acid within 2 weeks of surgery and only 47 had received placebo, and they saw no advantage to intravenous zoledronic acid compared with placebo in these first 2 weeks with respect to bone mineral density, fracture risk, or risk of death. However, excluding this small subset, antifracture efficacy and reduction in mortality rate were present when patients were treated with zoledronic acid in the 2 to 12 weeks after hip fracture repair, and improvement in bone mineral density at the hip was noted at 12 months in all cohorts.
Colón-Emeric et al3 performed another post hoc analysis, attempting to explain the lower mortality rate seen in patients treated with zoledronic acid. It had been an unexpected finding, and determinants of mortality rate reduction were hampered by a limited knowledge of the true cause of death or the circumstances of care after fracture. The authors concluded that only 8% of the reduction in mortality rate evident early in the second year of treatment with zoledronic acid could be attributed to a reduction in fractures.3 Other mechanisms by which the mortality rate reduction occurred remained unclear.
Curiously, in another large randomized controlled trial of zoledronic acid, in women with postmenopausal osteoporosis, Black et al4 reported that more patients died in the treated group (130 of 3,862) than in the placebo group (112 of 3,852). This difference was not statistically significant, but neither was it explained.
A meta-analysis by Bolland et al5 examined the effect of other osteoporosis treatments on mortality rate, using randomized controlled trials that lasted more than 12 months and that reported more than 10 deaths. The authors concluded the following:
In the trials in which bisphosphonates reduced the mortality rate, the mortality rate in the placebo group was higher than 10 per 1,000 patient-years
The effect of osteoporosis treatment on the mortality rate in a frail, elderly population is evident using agents with proven efficacy in reducing vertebral and nonvertebral fractures, eg, alendronate, risedronate, and zoledronic acid.5
THE SCIENCE
Osteoporotic fractures occur with minimal trauma, with the failure of bone attributed to impaired integrity of bone microarchitecture. The ultimate goal of fracture repair is to restore bone size, shape, and tissue properties. The issue of when to treat with a bisphosphonate after hip fracture arises because bisphosphonates are known to disrupt bone remodeling and so delay fracture repair.
After fracture, both anabolic and catabolic phases occur.6 The final outcome depends on the following:
The type of intervention to stabilize the fracture site (eg, surgical repair)
The inflammatory cytokines and growth factors released by the cellular elements in bloody and disrupted tissue.
Oxygen tension, angiogenesis, and osteoblasts are critical to primary bone formation, and osteoclasts are essential in remodeling this initial bone deposition. These late phases of fracture repair are most vulnerable to the bisphosphonates, through suppression of osteoclast resorption and possibly through decreased angiogenesis.6 Callus formation is sustained, but bone remodeling is delayed.
Amanat et al7 examined the timing of a single dose of zoledronic acid after fracture repair in a rat model of diaphyseal fracture and found that the callus was larger and stronger if the bisphosphonate dose had been delayed 1 or 2 weeks. The animals treated with zoledronic acid showed a remarkable trabecular network of bone between the original femoral cortex and the new cortical bone that was not present in the control group, perhaps contributing to the enhanced mechanical properties of the callus. Other studies suggest single dosing rather than continuous dosing may be advantageous in fracture healing.8
THE REALITY
Healthy dogs or growing rats with linear diaphyseal fractures are imperfect models for elderly osteoporotic patients with hip fracture, as Dr. Herbert Fleisch noted in his editorial, “Can bisphosphonates be given to patients with fractures?”9 Still, if retained primary bone can be used in the process of fracture repair to gain an early mechanical advantage, then perhaps delayed remodeling will permit early mobilization and further fracture prevention in humans.
How soon after hip fracture surgery should a patient start a bisphosphonate? The only data we have are from a single randomized controlled trial designed to measure fracture risk reduction in osteoporotic patients with hip fracture using intravenous zoledronic acid 5 mg compared with placebo.2 A post hoc analysis of this study1 generated the limited clinical data we have on the optimal timing of the treatment. Linking these study data with the laboratory data, one would intuit that delaying the infusion of zoledronic acid for at least 2 weeks after hip fracture repair would offer a clinical reduction in fracture risk and improvement (or stabilization) in bone mineral density by 12 months, and a reduction in the rate of all-cause mortality beginning at 16 months.
References
Eriksen EF, Lyles KW, Colón-Emeric CS, et al. Antifracture efficacy and reduction of mortality in relation to timing of the first dose of zoledronic acid after hip fracture. J Bone Miner Res2009; 24:1308–1313.
Lyles KW, Colón-Emeric CS, Magaziner JS, et al; for the HORIZON Recurrent Fracture Trial. Zoledronic acid and clinical fractures and mortality after hip fracture. N Engl J Med2007; 357:1799–1809.
Colón-Emeric CS, Mesenbrink P, Lyles KW, et al. Potential mediators of the mortality reduction with zoledronic acid after hip fracture. J Bone Miner Res2010; 25:91–97.
Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med2007; 356:1809–1822.
Bolland MJ, Grey AB, Gamble GD, Reid IR. Effect of osteoporosis treatment on mortality: a meta-analysis. J Clin Endocrinol Metab2010; 95:1174–1181.
Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: the cellular picture. Semin Cell Dev Biol2008; 19:459–466.
Amanat N, McDonald M, Godfrey C, Bilston L, Little D. Optimal timing of a single dose of zoledronic acid to increase strength in rat fracture repair. J Bone Miner Res2007; 22:867–876.
Li J, Mori S, Kaji Y, Mashiba T, Kawanishi J, Norimatsu H. Effect of bisphosphonate (incadronate) on fracture healing of long bones in rats. J Bone Miner Res1999; 14:969–979.
Fleisch H. Can bisphosphonates be given to patients with fractures?J Bone Miner Res2001; 16:437–440.
References
Eriksen EF, Lyles KW, Colón-Emeric CS, et al. Antifracture efficacy and reduction of mortality in relation to timing of the first dose of zoledronic acid after hip fracture. J Bone Miner Res2009; 24:1308–1313.
Lyles KW, Colón-Emeric CS, Magaziner JS, et al; for the HORIZON Recurrent Fracture Trial. Zoledronic acid and clinical fractures and mortality after hip fracture. N Engl J Med2007; 357:1799–1809.
Colón-Emeric CS, Mesenbrink P, Lyles KW, et al. Potential mediators of the mortality reduction with zoledronic acid after hip fracture. J Bone Miner Res2010; 25:91–97.
Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med2007; 356:1809–1822.
Bolland MJ, Grey AB, Gamble GD, Reid IR. Effect of osteoporosis treatment on mortality: a meta-analysis. J Clin Endocrinol Metab2010; 95:1174–1181.
Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: the cellular picture. Semin Cell Dev Biol2008; 19:459–466.
Amanat N, McDonald M, Godfrey C, Bilston L, Little D. Optimal timing of a single dose of zoledronic acid to increase strength in rat fracture repair. J Bone Miner Res2007; 22:867–876.
Li J, Mori S, Kaji Y, Mashiba T, Kawanishi J, Norimatsu H. Effect of bisphosphonate (incadronate) on fracture healing of long bones in rats. J Bone Miner Res1999; 14:969–979.
Fleisch H. Can bisphosphonates be given to patients with fractures?J Bone Miner Res2001; 16:437–440.
Although we often approach anticoagulation therapy with a confidence born of familiarity, it is not for the faint of heart. We start chronic anticoagulation in several clinical settings, such as to prevent a recurrence after a thromboembolic event. But this decision requires weighing the increased risk of bleeding from the anticoagulant therapy against the risk of another thromboembolic event.
Along with massive pulmonary embolism, the most feared thromboembolic event is the clot that migrates to the brain, resulting in life-altering stroke. We assess this risk in a semiquantitative manner in patients with atrial fibrillation using the CHADS2 score, hoping to maximize the benefits of anticoagulation while reducing the risks. We recognize that patients at the greatest risk of stroke in this setting are those with a history of a prior stroke. Also, patients bedridden with a recent cerebrovascular accident (CVA) seem to be hypercoagulable, potentially adding risk to recent injury. Thus, we try to start anticoagulation as soon as feasible after the diagnosis of a possible thrombotic event.
But the decision to start or resume anticoagulation is especially agonizing in a patient who has suffered an intracerebral hemorrhage. In this issue of the Journal, Drs. Joshua Goldstein and Steven Greenberg and Dr. Franklin Michota provide a thoughtful discussion of the issues we need to consider in these patients.
While not contributing to the prevention of additional CVAs or other arterial thrombotic events, a modality often underused in the prevention of thrombotic disease is the application (not just the ordering) of compressive leg stockings to bedridden hospitalized patients who cannot, for any reason, be provided pharmacologic anticoagulation therapy. I just completed a stint of hospital consultation, and I was pleased to see the widespread integration of prophylactic anticoagulation therapy, but somewhat dismayed by the number of compressive stockings I watched pumping with vigor, but to no one’s benefit, as they were draped over a bed rail.
As we struggle with complex clinical decisions, we need to also be attentive to the simple and the seemingly mundane: using the foam dispenser at the door, offering the verbal greeting and patient touch at the bedside, and rewrapping the pneumatic stockings that have somehow migrated between mattress and footboard.
Although we often approach anticoagulation therapy with a confidence born of familiarity, it is not for the faint of heart. We start chronic anticoagulation in several clinical settings, such as to prevent a recurrence after a thromboembolic event. But this decision requires weighing the increased risk of bleeding from the anticoagulant therapy against the risk of another thromboembolic event.
Along with massive pulmonary embolism, the most feared thromboembolic event is the clot that migrates to the brain, resulting in life-altering stroke. We assess this risk in a semiquantitative manner in patients with atrial fibrillation using the CHADS2 score, hoping to maximize the benefits of anticoagulation while reducing the risks. We recognize that patients at the greatest risk of stroke in this setting are those with a history of a prior stroke. Also, patients bedridden with a recent cerebrovascular accident (CVA) seem to be hypercoagulable, potentially adding risk to recent injury. Thus, we try to start anticoagulation as soon as feasible after the diagnosis of a possible thrombotic event.
But the decision to start or resume anticoagulation is especially agonizing in a patient who has suffered an intracerebral hemorrhage. In this issue of the Journal, Drs. Joshua Goldstein and Steven Greenberg and Dr. Franklin Michota provide a thoughtful discussion of the issues we need to consider in these patients.
While not contributing to the prevention of additional CVAs or other arterial thrombotic events, a modality often underused in the prevention of thrombotic disease is the application (not just the ordering) of compressive leg stockings to bedridden hospitalized patients who cannot, for any reason, be provided pharmacologic anticoagulation therapy. I just completed a stint of hospital consultation, and I was pleased to see the widespread integration of prophylactic anticoagulation therapy, but somewhat dismayed by the number of compressive stockings I watched pumping with vigor, but to no one’s benefit, as they were draped over a bed rail.
As we struggle with complex clinical decisions, we need to also be attentive to the simple and the seemingly mundane: using the foam dispenser at the door, offering the verbal greeting and patient touch at the bedside, and rewrapping the pneumatic stockings that have somehow migrated between mattress and footboard.
Although we often approach anticoagulation therapy with a confidence born of familiarity, it is not for the faint of heart. We start chronic anticoagulation in several clinical settings, such as to prevent a recurrence after a thromboembolic event. But this decision requires weighing the increased risk of bleeding from the anticoagulant therapy against the risk of another thromboembolic event.
Along with massive pulmonary embolism, the most feared thromboembolic event is the clot that migrates to the brain, resulting in life-altering stroke. We assess this risk in a semiquantitative manner in patients with atrial fibrillation using the CHADS2 score, hoping to maximize the benefits of anticoagulation while reducing the risks. We recognize that patients at the greatest risk of stroke in this setting are those with a history of a prior stroke. Also, patients bedridden with a recent cerebrovascular accident (CVA) seem to be hypercoagulable, potentially adding risk to recent injury. Thus, we try to start anticoagulation as soon as feasible after the diagnosis of a possible thrombotic event.
But the decision to start or resume anticoagulation is especially agonizing in a patient who has suffered an intracerebral hemorrhage. In this issue of the Journal, Drs. Joshua Goldstein and Steven Greenberg and Dr. Franklin Michota provide a thoughtful discussion of the issues we need to consider in these patients.
While not contributing to the prevention of additional CVAs or other arterial thrombotic events, a modality often underused in the prevention of thrombotic disease is the application (not just the ordering) of compressive leg stockings to bedridden hospitalized patients who cannot, for any reason, be provided pharmacologic anticoagulation therapy. I just completed a stint of hospital consultation, and I was pleased to see the widespread integration of prophylactic anticoagulation therapy, but somewhat dismayed by the number of compressive stockings I watched pumping with vigor, but to no one’s benefit, as they were draped over a bed rail.
As we struggle with complex clinical decisions, we need to also be attentive to the simple and the seemingly mundane: using the foam dispenser at the door, offering the verbal greeting and patient touch at the bedside, and rewrapping the pneumatic stockings that have somehow migrated between mattress and footboard.
Anticoagulants have been helping patients at risk of thrombosis since the late 1930s.1,2 Although the indications for these agents are many, the development of anticoagulants beyond oral vitamin K antagonists and parenteral heparin has been slow. In the United States, the vitamin K antagonist warfarin (Coumadin) is still the only oral anticoagulant available.
The major complication of anticoagulant therapy is bleeding, and vitamin K antagonists have proven challenging to use in clinical practice.1,3 They have a narrow therapeutic window, they vary considerably in dose-response from patient to patient, and they are subject to significant interactions with other drugs and with foods. For these reasons, therapy must be monitored with laboratory testing, and good patient compliance and patient education are essential. Yet even with these measures, life-threatening hemorrhage still can occur.
In this issue of the Cleveland Clinic Journal of Medicine, Goldstein and Greenberg4 review warfarin-related intracerebral hemorrhage (ICH) and provide a framework for considering whether to resume anticoagulant therapy.
WHAT TO DO IN THE ACUTE PHASE
Goldstein and Greenberg divide the difficult clinical question of what to do after ICH into the acute phase and the chronic phase.
What to do in the acute phase appears straightforward, as the risk of hematoma expansion in the hours immediately after warfarin-related ICH outweighs the risk of arterial or venous thromboembolism. Anticoagulant reversal should be the primary consideration in the first 24 hours, and, assuming the patient does not have acute (< 4-week-old) deep vein thrombosis, intermittent pneumatic compression should be applied to the lower extremities to reduce the risk of venous thromboembolism associated with ICH.5
Prophylactic anticoagulation with subcutaneous fixed-dose heparin or low-molecular-weight heparin is recommended starting 72 hours after ICH is diagnosed, provided the patient is not underweight (< 50 kg), has relatively normal renal function (creatinine clearance > 30 mL/minute/1.73 m2) and normal platelet function, and does not have coagulopathy. 6 If any one of these criteria is not met, the risk of bleeding can be higher, even with only prophylactic doses of anticoagulant drugs. Prophylactic anticoagulation should be continued until hospital and rehabilitation discharge, typically 1 to 2 weeks after ICH, depending on the severity of the patient’s neurologic impairment.
If a patient with warfarin-related ICH has concomitant acute proximal deep vein thrombosis or pulmonary embolism (ie, < 4 weeks old), then caval interruption therapy would be indicated.7 Although retrievable inferior vena cava filters are increasingly preferred over permanent filters, it is important to recognize the relative lack of both longitudinal and prospective data on retrievable devices. Given that provoked venous thromboembolism requires a minimum of 3 months of anticoagulation, and retrievable filters generally need to be removed before 3 months, a retrievable filter should be chosen only if the clinician has already decided that oral anticoagulation will be restarted in the next 3 to 4 weeks after filter removal.
WHAT TO DO IN THE CHRONIC PHASE
A more difficult question in patients with warfarin-related ICH arises in the chronic phase: should oral anticoagulation be resumed at all?
Goldstein and Greenberg outline important considerations. Under the principle of primum non nocere, patients who have suffered a warfarin-related ICH should first be evaluated for their risk of thrombosis in light of their original indication for oral anticoagulant therapy. As the authors point out, oral anticoagulation for primary prevention of thrombosis after warfarin-related ICH must be viewed differently than oral anticoagulation for secondary prevention of thrombosis. In addition, Douketis et al8 have described a method of stratifying a patient’s risk of thrombosis as low, moderate, or high (Table 1), which is the basis for decisions about perioperative anticoagulation. Based on Goldstein and Greenberg’s review, we can similarly categorize these patients as being at low, moderate, or high risk of ICH recurrence (Table 2). Patients at low risk of thrombosis should probably not resume taking a vitamin K antagonist, regardless of their ICH risk (Table 3). It would be reasonable, however, for patients at moderate or high risk of thrombosis and at low risk of ICH to resume taking their vitamin K antagonist.
Uncertainty remains for patients with a moderate or high risk of thrombosis and a moderate or high risk of ICH. For patients with these combinations of risk, individualized approaches need to be explored. All attempts should be made to widen the margin of safety of vitamin K antagonist therapy; these include referring the patient to an anticoagulation management service, frequent laboratory monitoring, and ongoing patient education.1
Since the risk of ICH is related to the intensity of anticoagulation, a lower target international normalized ratio may be the best compromise, depending on the patient. Alternatively, antiplatelet therapy alone may offer some benefit with less risk of ICH.
THE NEWER ORAL ANTICOAGULANTS
As Goldstein and Greenberg mention, the ongoing development of new and potentially safer oral anticoagulants may affect how we approach these risk-benefit equations.
Three new oral anticoagulants—dabigatran (Pradaxa), apixaban, and rivaroxaban (Xarelto)—are being tested for various anticoagulant indications, and several phase III studies have recently closed or are nearing completion.
Dabigatran is an oral direct thrombin inhibitor currently available in Europe and Canada.
In the Randomized Evaluation of Long-term Anticoagulant Therapy (RE-LY) trial, the efficacy and safety of two different doses of dabigatran (110 mg twice daily or 150 mg twice daily) relative to warfarin were studied in more than 18,000 patients with atrial fibrillation. 9 The primary outcome measure was stroke or systemic embolism. Dabigatran 110 mg was not inferior to warfarin in terms of the primary outcome, while dabigatran 150 mg was superior. The rate of major bleeding was 3.36% per year in the warfarin group vs 2.71% in the 110-mg group (P = .003) and 3.11% in the 150-mg group (P not significant).
Additional safety data on this drug are available from the 2,500-patient RE-COVER trial.10 Dabigatran was not inferior to warfarin in the treatment of acute venous thromboembolism, with a similar rate of major bleeding and a lower rate of combined major plus nonmajor bleeding.
Apixaban, an oral direct factor Xa inhibitor, is in a phase III trial in patients with atrial fibrillation—Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE)11—comparing apixaban vs warfarin. Another phase III trial, AVERROES,12 was stopped early after a predefined interim analysis by the independent data-monitoring committee found clear evidence of benefit in the apixaban group.13 The AVERROES results were presented at the 2010 European Society of Cardiology Congress, August 28–September 1, Stockholm, Sweden.14
Rivaroxaban, another promising oral direct factor Xa inhibitor, is currently available in Europe and Canada for the prevention of thrombosis in orthopedic surgery patients. Rivaroxaban is also in large phase III trials for the treatment of acute venous thromboembolism15–17 and for the prevention of stroke in atrial fibrillation.18
Newer agents have drawbacks, too
These new agents need no laboratory monitoring, and they do not appear to be subject to the dose variability and the interactions with drugs and foods seen with vitamin K antagonists. As a result, they may pose less risk of anticoagulant-related ICH.
The decision to resume anticoagulation after anticoagulant-associated intracranial hemorrhage should be based on the risk of rebleeding vs the risk of thrombosis. Patients determined to be at high risk of thrombosis and low risk of rebleeding are the best candidates for resuming anticoagulation.
Still, for patients who suffer an anticoagulant- or warfarin-related ICH, these new anticoagulants are not likely to simplify the issue of restarting anticoagulant therapy. Unlike vitamin K antagonists, dabigatran and the direct factor Xa inhibitors have no known antidote for their anticoagulant effects. Animal data suggest that factor Xa concentrates may help,19 but for patients at risk of a second anticoagulant-related ICH, this does not provide much reassurance.
As with all clinical decisions in medicine, the potential benefits of any therapy should outweigh the risks. In the case of warfarin-related ICH, resuming anticoagulant therapy requires careful consideration of many factors, including patient preferences and tolerance of different levels of risk. As new and perhaps safer anticoagulants become available, clinicians may face such difficult questions less and less. But in the meantime, doctors and their patients are left to pick their poison.
References
Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G; American College of Chest Physicians. Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):160S–198S.
Hirsh J, Bauer KA, Donati MB, Gould M, Samama MM, Weitz JI; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):141S–159S.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th edition). Chest2008; 133(suppl 6):257S–298S.
Goldstein JN, Greenberg SM. Should anticoagulation be resumed after intracerebral hemorrhage?Cleve Clin J Med2010; 77:791–799.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Michota F, Merli G. Anticoagulation in special patient populations: are special dosing considerations required?Cleve Clin J Med2005; 72(suppl 1):S37–S42.
Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE, Comerota AJ; American College of Chest Physicians. Antithrombotic therapy for venous thromboembolic disease: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):454S–545S.
Douketis JD, Berger PB, Dunn AS, et al; American College of Chest Physicians. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):299S–339S.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Lopes RD, Alexander JH, Al-Khatib SM; ARISTOTLE Investigators. Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial: design and rationale. Am Heart J2010; 159:331–339.
Eikelboom JW, O’Donnell M, Yusuf S, et al. Rationale and design of AVERROES: apixaban versus acetylsalicylic acid to prevent stroke in atrial fibrillation patients who have failed or are unsuitable for vitamin K antagonist treatment. Am Heart J2010; 159:348–353.
Pfizer/Bristol-Myers Squibb. AVERROES study of investigational agent apixaban closes early due to clear evidence of efficacy, June 9, 2010. www.theheart.org/article/1087291.do. Accessed September 26, 2010.
Once-daily oral direct factor Xa inhibitor rivaroxaban in the long-term prevention of recurrent symptomatic venous thromboembolism in patients with symptomatic deep-vein thrombosis or pulmonary embolism. The Einstein-Extension Study. http://clinicaltrials.gov/ct2/show/NCT00439725. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic deep-vein thrombosis without symptomatic pulmonary embolism: Einstein-DVT Evaluation. http://clinicaltrials.gov/ct2/show/NCT00440193. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic pulmonary embolism with or without symptomatic deep-vein thrombosis: Einstein-PE Evaluation. http://clinicaltrials.gov/ct2/show/NCT00439777. Accessed September 26, 2010.
ROCKET AF Study Investigators. Rivaroxaban once-daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation: rationale and design of the ROCKET AF study. Am Heart J2010; 159:340–347.
Weitz JI, Hirsh J, Samama MM; American College of Chest Physicians. New antithrombotic drugs: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):234S–256S.
Franklin Michota, MD, FHM Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, and Director of Academic Affairs, Department of Hospital Medicine, Cleveland Clinic
Address: Franklin Michota, MD, Department of Hospital Medicine, M2 Anx, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Franklin Michota, MD, FHM Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, and Director of Academic Affairs, Department of Hospital Medicine, Cleveland Clinic
Address: Franklin Michota, MD, Department of Hospital Medicine, M2 Anx, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Author and Disclosure Information
Franklin Michota, MD, FHM Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, and Director of Academic Affairs, Department of Hospital Medicine, Cleveland Clinic
Address: Franklin Michota, MD, Department of Hospital Medicine, M2 Anx, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]
Anticoagulants have been helping patients at risk of thrombosis since the late 1930s.1,2 Although the indications for these agents are many, the development of anticoagulants beyond oral vitamin K antagonists and parenteral heparin has been slow. In the United States, the vitamin K antagonist warfarin (Coumadin) is still the only oral anticoagulant available.
The major complication of anticoagulant therapy is bleeding, and vitamin K antagonists have proven challenging to use in clinical practice.1,3 They have a narrow therapeutic window, they vary considerably in dose-response from patient to patient, and they are subject to significant interactions with other drugs and with foods. For these reasons, therapy must be monitored with laboratory testing, and good patient compliance and patient education are essential. Yet even with these measures, life-threatening hemorrhage still can occur.
In this issue of the Cleveland Clinic Journal of Medicine, Goldstein and Greenberg4 review warfarin-related intracerebral hemorrhage (ICH) and provide a framework for considering whether to resume anticoagulant therapy.
WHAT TO DO IN THE ACUTE PHASE
Goldstein and Greenberg divide the difficult clinical question of what to do after ICH into the acute phase and the chronic phase.
What to do in the acute phase appears straightforward, as the risk of hematoma expansion in the hours immediately after warfarin-related ICH outweighs the risk of arterial or venous thromboembolism. Anticoagulant reversal should be the primary consideration in the first 24 hours, and, assuming the patient does not have acute (< 4-week-old) deep vein thrombosis, intermittent pneumatic compression should be applied to the lower extremities to reduce the risk of venous thromboembolism associated with ICH.5
Prophylactic anticoagulation with subcutaneous fixed-dose heparin or low-molecular-weight heparin is recommended starting 72 hours after ICH is diagnosed, provided the patient is not underweight (< 50 kg), has relatively normal renal function (creatinine clearance > 30 mL/minute/1.73 m2) and normal platelet function, and does not have coagulopathy. 6 If any one of these criteria is not met, the risk of bleeding can be higher, even with only prophylactic doses of anticoagulant drugs. Prophylactic anticoagulation should be continued until hospital and rehabilitation discharge, typically 1 to 2 weeks after ICH, depending on the severity of the patient’s neurologic impairment.
If a patient with warfarin-related ICH has concomitant acute proximal deep vein thrombosis or pulmonary embolism (ie, < 4 weeks old), then caval interruption therapy would be indicated.7 Although retrievable inferior vena cava filters are increasingly preferred over permanent filters, it is important to recognize the relative lack of both longitudinal and prospective data on retrievable devices. Given that provoked venous thromboembolism requires a minimum of 3 months of anticoagulation, and retrievable filters generally need to be removed before 3 months, a retrievable filter should be chosen only if the clinician has already decided that oral anticoagulation will be restarted in the next 3 to 4 weeks after filter removal.
WHAT TO DO IN THE CHRONIC PHASE
A more difficult question in patients with warfarin-related ICH arises in the chronic phase: should oral anticoagulation be resumed at all?
Goldstein and Greenberg outline important considerations. Under the principle of primum non nocere, patients who have suffered a warfarin-related ICH should first be evaluated for their risk of thrombosis in light of their original indication for oral anticoagulant therapy. As the authors point out, oral anticoagulation for primary prevention of thrombosis after warfarin-related ICH must be viewed differently than oral anticoagulation for secondary prevention of thrombosis. In addition, Douketis et al8 have described a method of stratifying a patient’s risk of thrombosis as low, moderate, or high (Table 1), which is the basis for decisions about perioperative anticoagulation. Based on Goldstein and Greenberg’s review, we can similarly categorize these patients as being at low, moderate, or high risk of ICH recurrence (Table 2). Patients at low risk of thrombosis should probably not resume taking a vitamin K antagonist, regardless of their ICH risk (Table 3). It would be reasonable, however, for patients at moderate or high risk of thrombosis and at low risk of ICH to resume taking their vitamin K antagonist.
Uncertainty remains for patients with a moderate or high risk of thrombosis and a moderate or high risk of ICH. For patients with these combinations of risk, individualized approaches need to be explored. All attempts should be made to widen the margin of safety of vitamin K antagonist therapy; these include referring the patient to an anticoagulation management service, frequent laboratory monitoring, and ongoing patient education.1
Since the risk of ICH is related to the intensity of anticoagulation, a lower target international normalized ratio may be the best compromise, depending on the patient. Alternatively, antiplatelet therapy alone may offer some benefit with less risk of ICH.
THE NEWER ORAL ANTICOAGULANTS
As Goldstein and Greenberg mention, the ongoing development of new and potentially safer oral anticoagulants may affect how we approach these risk-benefit equations.
Three new oral anticoagulants—dabigatran (Pradaxa), apixaban, and rivaroxaban (Xarelto)—are being tested for various anticoagulant indications, and several phase III studies have recently closed or are nearing completion.
Dabigatran is an oral direct thrombin inhibitor currently available in Europe and Canada.
In the Randomized Evaluation of Long-term Anticoagulant Therapy (RE-LY) trial, the efficacy and safety of two different doses of dabigatran (110 mg twice daily or 150 mg twice daily) relative to warfarin were studied in more than 18,000 patients with atrial fibrillation. 9 The primary outcome measure was stroke or systemic embolism. Dabigatran 110 mg was not inferior to warfarin in terms of the primary outcome, while dabigatran 150 mg was superior. The rate of major bleeding was 3.36% per year in the warfarin group vs 2.71% in the 110-mg group (P = .003) and 3.11% in the 150-mg group (P not significant).
Additional safety data on this drug are available from the 2,500-patient RE-COVER trial.10 Dabigatran was not inferior to warfarin in the treatment of acute venous thromboembolism, with a similar rate of major bleeding and a lower rate of combined major plus nonmajor bleeding.
Apixaban, an oral direct factor Xa inhibitor, is in a phase III trial in patients with atrial fibrillation—Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE)11—comparing apixaban vs warfarin. Another phase III trial, AVERROES,12 was stopped early after a predefined interim analysis by the independent data-monitoring committee found clear evidence of benefit in the apixaban group.13 The AVERROES results were presented at the 2010 European Society of Cardiology Congress, August 28–September 1, Stockholm, Sweden.14
Rivaroxaban, another promising oral direct factor Xa inhibitor, is currently available in Europe and Canada for the prevention of thrombosis in orthopedic surgery patients. Rivaroxaban is also in large phase III trials for the treatment of acute venous thromboembolism15–17 and for the prevention of stroke in atrial fibrillation.18
Newer agents have drawbacks, too
These new agents need no laboratory monitoring, and they do not appear to be subject to the dose variability and the interactions with drugs and foods seen with vitamin K antagonists. As a result, they may pose less risk of anticoagulant-related ICH.
The decision to resume anticoagulation after anticoagulant-associated intracranial hemorrhage should be based on the risk of rebleeding vs the risk of thrombosis. Patients determined to be at high risk of thrombosis and low risk of rebleeding are the best candidates for resuming anticoagulation.
Still, for patients who suffer an anticoagulant- or warfarin-related ICH, these new anticoagulants are not likely to simplify the issue of restarting anticoagulant therapy. Unlike vitamin K antagonists, dabigatran and the direct factor Xa inhibitors have no known antidote for their anticoagulant effects. Animal data suggest that factor Xa concentrates may help,19 but for patients at risk of a second anticoagulant-related ICH, this does not provide much reassurance.
As with all clinical decisions in medicine, the potential benefits of any therapy should outweigh the risks. In the case of warfarin-related ICH, resuming anticoagulant therapy requires careful consideration of many factors, including patient preferences and tolerance of different levels of risk. As new and perhaps safer anticoagulants become available, clinicians may face such difficult questions less and less. But in the meantime, doctors and their patients are left to pick their poison.
Anticoagulants have been helping patients at risk of thrombosis since the late 1930s.1,2 Although the indications for these agents are many, the development of anticoagulants beyond oral vitamin K antagonists and parenteral heparin has been slow. In the United States, the vitamin K antagonist warfarin (Coumadin) is still the only oral anticoagulant available.
The major complication of anticoagulant therapy is bleeding, and vitamin K antagonists have proven challenging to use in clinical practice.1,3 They have a narrow therapeutic window, they vary considerably in dose-response from patient to patient, and they are subject to significant interactions with other drugs and with foods. For these reasons, therapy must be monitored with laboratory testing, and good patient compliance and patient education are essential. Yet even with these measures, life-threatening hemorrhage still can occur.
In this issue of the Cleveland Clinic Journal of Medicine, Goldstein and Greenberg4 review warfarin-related intracerebral hemorrhage (ICH) and provide a framework for considering whether to resume anticoagulant therapy.
WHAT TO DO IN THE ACUTE PHASE
Goldstein and Greenberg divide the difficult clinical question of what to do after ICH into the acute phase and the chronic phase.
What to do in the acute phase appears straightforward, as the risk of hematoma expansion in the hours immediately after warfarin-related ICH outweighs the risk of arterial or venous thromboembolism. Anticoagulant reversal should be the primary consideration in the first 24 hours, and, assuming the patient does not have acute (< 4-week-old) deep vein thrombosis, intermittent pneumatic compression should be applied to the lower extremities to reduce the risk of venous thromboembolism associated with ICH.5
Prophylactic anticoagulation with subcutaneous fixed-dose heparin or low-molecular-weight heparin is recommended starting 72 hours after ICH is diagnosed, provided the patient is not underweight (< 50 kg), has relatively normal renal function (creatinine clearance > 30 mL/minute/1.73 m2) and normal platelet function, and does not have coagulopathy. 6 If any one of these criteria is not met, the risk of bleeding can be higher, even with only prophylactic doses of anticoagulant drugs. Prophylactic anticoagulation should be continued until hospital and rehabilitation discharge, typically 1 to 2 weeks after ICH, depending on the severity of the patient’s neurologic impairment.
If a patient with warfarin-related ICH has concomitant acute proximal deep vein thrombosis or pulmonary embolism (ie, < 4 weeks old), then caval interruption therapy would be indicated.7 Although retrievable inferior vena cava filters are increasingly preferred over permanent filters, it is important to recognize the relative lack of both longitudinal and prospective data on retrievable devices. Given that provoked venous thromboembolism requires a minimum of 3 months of anticoagulation, and retrievable filters generally need to be removed before 3 months, a retrievable filter should be chosen only if the clinician has already decided that oral anticoagulation will be restarted in the next 3 to 4 weeks after filter removal.
WHAT TO DO IN THE CHRONIC PHASE
A more difficult question in patients with warfarin-related ICH arises in the chronic phase: should oral anticoagulation be resumed at all?
Goldstein and Greenberg outline important considerations. Under the principle of primum non nocere, patients who have suffered a warfarin-related ICH should first be evaluated for their risk of thrombosis in light of their original indication for oral anticoagulant therapy. As the authors point out, oral anticoagulation for primary prevention of thrombosis after warfarin-related ICH must be viewed differently than oral anticoagulation for secondary prevention of thrombosis. In addition, Douketis et al8 have described a method of stratifying a patient’s risk of thrombosis as low, moderate, or high (Table 1), which is the basis for decisions about perioperative anticoagulation. Based on Goldstein and Greenberg’s review, we can similarly categorize these patients as being at low, moderate, or high risk of ICH recurrence (Table 2). Patients at low risk of thrombosis should probably not resume taking a vitamin K antagonist, regardless of their ICH risk (Table 3). It would be reasonable, however, for patients at moderate or high risk of thrombosis and at low risk of ICH to resume taking their vitamin K antagonist.
Uncertainty remains for patients with a moderate or high risk of thrombosis and a moderate or high risk of ICH. For patients with these combinations of risk, individualized approaches need to be explored. All attempts should be made to widen the margin of safety of vitamin K antagonist therapy; these include referring the patient to an anticoagulation management service, frequent laboratory monitoring, and ongoing patient education.1
Since the risk of ICH is related to the intensity of anticoagulation, a lower target international normalized ratio may be the best compromise, depending on the patient. Alternatively, antiplatelet therapy alone may offer some benefit with less risk of ICH.
THE NEWER ORAL ANTICOAGULANTS
As Goldstein and Greenberg mention, the ongoing development of new and potentially safer oral anticoagulants may affect how we approach these risk-benefit equations.
Three new oral anticoagulants—dabigatran (Pradaxa), apixaban, and rivaroxaban (Xarelto)—are being tested for various anticoagulant indications, and several phase III studies have recently closed or are nearing completion.
Dabigatran is an oral direct thrombin inhibitor currently available in Europe and Canada.
In the Randomized Evaluation of Long-term Anticoagulant Therapy (RE-LY) trial, the efficacy and safety of two different doses of dabigatran (110 mg twice daily or 150 mg twice daily) relative to warfarin were studied in more than 18,000 patients with atrial fibrillation. 9 The primary outcome measure was stroke or systemic embolism. Dabigatran 110 mg was not inferior to warfarin in terms of the primary outcome, while dabigatran 150 mg was superior. The rate of major bleeding was 3.36% per year in the warfarin group vs 2.71% in the 110-mg group (P = .003) and 3.11% in the 150-mg group (P not significant).
Additional safety data on this drug are available from the 2,500-patient RE-COVER trial.10 Dabigatran was not inferior to warfarin in the treatment of acute venous thromboembolism, with a similar rate of major bleeding and a lower rate of combined major plus nonmajor bleeding.
Apixaban, an oral direct factor Xa inhibitor, is in a phase III trial in patients with atrial fibrillation—Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE)11—comparing apixaban vs warfarin. Another phase III trial, AVERROES,12 was stopped early after a predefined interim analysis by the independent data-monitoring committee found clear evidence of benefit in the apixaban group.13 The AVERROES results were presented at the 2010 European Society of Cardiology Congress, August 28–September 1, Stockholm, Sweden.14
Rivaroxaban, another promising oral direct factor Xa inhibitor, is currently available in Europe and Canada for the prevention of thrombosis in orthopedic surgery patients. Rivaroxaban is also in large phase III trials for the treatment of acute venous thromboembolism15–17 and for the prevention of stroke in atrial fibrillation.18
Newer agents have drawbacks, too
These new agents need no laboratory monitoring, and they do not appear to be subject to the dose variability and the interactions with drugs and foods seen with vitamin K antagonists. As a result, they may pose less risk of anticoagulant-related ICH.
The decision to resume anticoagulation after anticoagulant-associated intracranial hemorrhage should be based on the risk of rebleeding vs the risk of thrombosis. Patients determined to be at high risk of thrombosis and low risk of rebleeding are the best candidates for resuming anticoagulation.
Still, for patients who suffer an anticoagulant- or warfarin-related ICH, these new anticoagulants are not likely to simplify the issue of restarting anticoagulant therapy. Unlike vitamin K antagonists, dabigatran and the direct factor Xa inhibitors have no known antidote for their anticoagulant effects. Animal data suggest that factor Xa concentrates may help,19 but for patients at risk of a second anticoagulant-related ICH, this does not provide much reassurance.
As with all clinical decisions in medicine, the potential benefits of any therapy should outweigh the risks. In the case of warfarin-related ICH, resuming anticoagulant therapy requires careful consideration of many factors, including patient preferences and tolerance of different levels of risk. As new and perhaps safer anticoagulants become available, clinicians may face such difficult questions less and less. But in the meantime, doctors and their patients are left to pick their poison.
References
Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G; American College of Chest Physicians. Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):160S–198S.
Hirsh J, Bauer KA, Donati MB, Gould M, Samama MM, Weitz JI; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):141S–159S.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th edition). Chest2008; 133(suppl 6):257S–298S.
Goldstein JN, Greenberg SM. Should anticoagulation be resumed after intracerebral hemorrhage?Cleve Clin J Med2010; 77:791–799.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Michota F, Merli G. Anticoagulation in special patient populations: are special dosing considerations required?Cleve Clin J Med2005; 72(suppl 1):S37–S42.
Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE, Comerota AJ; American College of Chest Physicians. Antithrombotic therapy for venous thromboembolic disease: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):454S–545S.
Douketis JD, Berger PB, Dunn AS, et al; American College of Chest Physicians. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):299S–339S.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Lopes RD, Alexander JH, Al-Khatib SM; ARISTOTLE Investigators. Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial: design and rationale. Am Heart J2010; 159:331–339.
Eikelboom JW, O’Donnell M, Yusuf S, et al. Rationale and design of AVERROES: apixaban versus acetylsalicylic acid to prevent stroke in atrial fibrillation patients who have failed or are unsuitable for vitamin K antagonist treatment. Am Heart J2010; 159:348–353.
Pfizer/Bristol-Myers Squibb. AVERROES study of investigational agent apixaban closes early due to clear evidence of efficacy, June 9, 2010. www.theheart.org/article/1087291.do. Accessed September 26, 2010.
Once-daily oral direct factor Xa inhibitor rivaroxaban in the long-term prevention of recurrent symptomatic venous thromboembolism in patients with symptomatic deep-vein thrombosis or pulmonary embolism. The Einstein-Extension Study. http://clinicaltrials.gov/ct2/show/NCT00439725. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic deep-vein thrombosis without symptomatic pulmonary embolism: Einstein-DVT Evaluation. http://clinicaltrials.gov/ct2/show/NCT00440193. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic pulmonary embolism with or without symptomatic deep-vein thrombosis: Einstein-PE Evaluation. http://clinicaltrials.gov/ct2/show/NCT00439777. Accessed September 26, 2010.
ROCKET AF Study Investigators. Rivaroxaban once-daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation: rationale and design of the ROCKET AF study. Am Heart J2010; 159:340–347.
Weitz JI, Hirsh J, Samama MM; American College of Chest Physicians. New antithrombotic drugs: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):234S–256S.
References
Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G; American College of Chest Physicians. Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):160S–198S.
Hirsh J, Bauer KA, Donati MB, Gould M, Samama MM, Weitz JI; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):141S–159S.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th edition). Chest2008; 133(suppl 6):257S–298S.
Goldstein JN, Greenberg SM. Should anticoagulation be resumed after intracerebral hemorrhage?Cleve Clin J Med2010; 77:791–799.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Michota F, Merli G. Anticoagulation in special patient populations: are special dosing considerations required?Cleve Clin J Med2005; 72(suppl 1):S37–S42.
Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE, Comerota AJ; American College of Chest Physicians. Antithrombotic therapy for venous thromboembolic disease: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):454S–545S.
Douketis JD, Berger PB, Dunn AS, et al; American College of Chest Physicians. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):299S–339S.
Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med2009; 361:1139–1151.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Lopes RD, Alexander JH, Al-Khatib SM; ARISTOTLE Investigators. Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial: design and rationale. Am Heart J2010; 159:331–339.
Eikelboom JW, O’Donnell M, Yusuf S, et al. Rationale and design of AVERROES: apixaban versus acetylsalicylic acid to prevent stroke in atrial fibrillation patients who have failed or are unsuitable for vitamin K antagonist treatment. Am Heart J2010; 159:348–353.
Pfizer/Bristol-Myers Squibb. AVERROES study of investigational agent apixaban closes early due to clear evidence of efficacy, June 9, 2010. www.theheart.org/article/1087291.do. Accessed September 26, 2010.
Once-daily oral direct factor Xa inhibitor rivaroxaban in the long-term prevention of recurrent symptomatic venous thromboembolism in patients with symptomatic deep-vein thrombosis or pulmonary embolism. The Einstein-Extension Study. http://clinicaltrials.gov/ct2/show/NCT00439725. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic deep-vein thrombosis without symptomatic pulmonary embolism: Einstein-DVT Evaluation. http://clinicaltrials.gov/ct2/show/NCT00440193. Accessed September 26, 2010.
Oral direct factor Xa inhibitor rivaroxaban in patients with acute symptomatic pulmonary embolism with or without symptomatic deep-vein thrombosis: Einstein-PE Evaluation. http://clinicaltrials.gov/ct2/show/NCT00439777. Accessed September 26, 2010.
ROCKET AF Study Investigators. Rivaroxaban once-daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation: rationale and design of the ROCKET AF study. Am Heart J2010; 159:340–347.
Weitz JI, Hirsh J, Samama MM; American College of Chest Physicians. New antithrombotic drugs: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):234S–256S.
If a patient taking warfarin (Coumadin) or other anticoagulant drug suffers an intracerebral hemorrhage (ICH) and survives, the physician faces the dilemma of whether to resume the anticoagulant. On one hand, the drug was prescribed because the patient was at risk of a thromboembolic event such as stroke or pulmonary embolism. On the other hand, warfarin use may increase the risk of another ICH.
Unfortunately, we have little evidence from clinical trials on which to base the decision. Nevertheless, we believe that in selected patients the potential benefit of resuming anticoagulation outweighs the considerable risk.
In the pages that follow, we summarize when and how anticoagulation therapy should be resumed after ICH.
A DEADLY COMPLICATION OF ANTICOAGULANT THERAPY
Intracranial bleeding is the most feared and the most deadly complication of oral anticoagulant therapy.1 The substantial risks associated with oral anticoagulants likely account for these drugs being underprescribed in patients who have indications for them.2–4
While bleeding is the major risk, not all bleeding events are equally damaging. Extracranial bleeding (eg, gastrointestinal bleeding, hematuria, epistaxis) leads to death or disability in only 3% of cases, whereas intracranial bleeding such as ICH leads to death or disability 76% of cases.5
Even without anticoagulation, ICH is the deadliest form of stroke,6–9 and if the patient has been taking warfarin, the risk of disability and death is substantially higher.6,10 Warfarin has a striking effect on the incidence and outcomes of ICH. While the overall incidence of ICH in the general population is approximately 25 per 100,000 person-years, the incidence in patients on warfarin is exponentially higher, at 2 to 3 per 100 per year, and appears to be increasing.11,12 In addition, once ICH occurs, the risk of death is up to twice as high in those on warfarin.6 The bulk of this effect is likely due to a higher risk of ongoing bleeding after the event.10,13–16
Major risk factors for ICH in patients taking oral anticoagulants include a higher international normalized ratio (INR) and older age.11,17
TWO KEY QUESTIONS
Once a patient is diagnosed with warfarin-related ICH, clinicians typically take urgent measures to restore normal coagulation, hoping to limit ongoing bleeding and improve outcome.18,19
The higher the INR at presentation, the greater the risk of death.6 In addition, in retrospective studies, some authors have noted that earlier correction of the INR is associated with better outcome.14,16
While emergency reversal of warfarin is widely considered standard treatment in the acute phase,20–24 concern persists about its safety in patients at high risk of thromboembolism.
Until the results of clinical trials are available, decisions about whether to reverse and when to resume anticoagulation hinge on two questions:
In the acute phase, how does the risk of further bleeding (hematoma expansion) compare with the short-term risk of thromboembolism?
In the chronic phase, how does the risk of recurrent hemorrhage compare with the excess risk of thromboembolism if the patient does not resume anticoagulation therapy?
ACUTELY, THE RISK OF BLEEDING OUTWEIGHS THAT OF CLOTTING
High risk of hematoma expansion after ICH
Unfortunately, continued bleeding is common after ICH. In patients who present within 3 hours of symptom onset, 26% of hematomas expand more than 33% over the first hour, and another 12% expand this amount over the next 20 hours.6 In warfarin-associated ICH, up to 50% of patients develop this level of hematoma expansion, but it appears to take place over a more prolonged period of time.10,13–16 Over 70% of patients presenting acutely develop at least some amount of expansion within 24 hours.25 Therefore, the risk of hematoma expansion in the first 24 hours is likely so high that patients cannot safely receive anticoagulants during this time frame.
But not all patients are at equal risk of hematoma expansion. Several features are associated with higher risk (Table 1)10,26–40:
A large hematoma volume on presentation is a significant predictor of expansion, possibly reflecting a more severe underlying insult.26,27
Early presentation, especially within 3 hours of symptom onset, also appears to mark those at higher risk, presumably because such patients undergo computed tomography (CT) while still bleeding.26,27
Figure 1. The “spot sign” (arrow), contrast extravasation after contrast-enhanced computed tomography, is associated with a high risk of hematoma expansion.For those on warfarin, a higher INR is a significant predictor, not just of higher risk, but also of a more delayed expansion.10,28
Certain radiographic findings indicate higher risk. One is the “spot sign,” ie, contrast extravasation after contrast-enhanced CT27,29–31 (Figure 1). Apparently, the more spots present, and the denser the contrast, the greater the risk, an observation that has led to a proposed “spot-sign score” that may predict both expansion and poor outcome. 32,41
Given the high risk of hematoma expansion in the early phase, and given our inability to predict hematoma expansion, most authorities recommend immediate reversal of anticoagulation after diagnosis.42–44 Reversal of anticoagulation typically includes intravenous vitamin K, which begins to act within several hours, and repletion of coagulation factors, which act within minutes (prothrombin complex concentrates and recombinant factor VIIa [NovoSeven]) or a few hours (fresh frozen plasma).1
Dosages:
Vitamin K 5 to 10 mg intravenously
Prothrombin complex concentrates 10 to 50 U/kg
Recombinant factor VIIa 40 to 80 μg/kg
Fresh frozen plasma 10 to 50 U/kg.
Risk of thromboembolism after ICH: Ongoing and cumulative
Thromboembolism after ICH is a major concern, for two main reasons.
First, patients on oral anticoagulation typically have a preexisting risk factor and are thus at higher risk of a thromboembolic event, particularly while they are off anticoagulation. Patients with atrial fibrillation or a mechanical valve are at risk of arterial events such as ischemic stroke, whereas patients with a known venous thromboembolic condition such as deep venous thrombosis or pulmonary embolism are at risk of extension of the thrombosis or recurrence of a venous thrombotic event.
Second, ICH itself increases the risk of arterial and venous thromboembolic events. Including patients not previously on anticoagulation, this risk is as high as 7% during the initial hospitalization and 9% during the first 90 days.45,46 Worth noting is that patients who previously received anticoagulant drugs (and who are off this therapy in the acute phase) are at no higher risk of thromboembolism compared with those who never received anticoagulants.45
However, while the risk of hematoma expansion is highest at presentation and then decreases with time, the risk of thromboembolism (particularly venous thromboembolism) is ongoing and cumulative. Arterial thromboembolism is more likely to occur early, within the first week, whereas venous thromboembolism can occur later.45
Overall, studies have estimated the short-term risk of pulmonary embolism to be 1% to 2%, deep venous thrombosis 1% to 4%, myocardial ischemia about 2%, and cerebral ischemia 2% to 3%.45,46 However, when patients are actively screened, the incidence of asymptomatic deep venous thrombosis is found to be as high as 16% in the first 10 days,47 and evidence of myocardial ischemia can be detected in up to 27% of patients.48
Therefore, the risk of hematoma expansion appears to be high and the risk of thromboembolism appears to be low during the first day after ICH. Over the next days, as the risk of hematoma expansion recedes, this ratio shifts.
Studies of in-hospital anticoagulation after ICH
The data on restarting oral anticoagulation in the acute phase are sparse. In practice, clinicians typically start heparinoids in low subcutaneous doses to prevent deep venous thrombosis and, after the first few days or a week, consider increasing to a full anticoagulation dose or starting an oral anticoagulant and subsequently discontinuing the heparin when the INR is in the therapeutic range (see discussion below).
ICH patients in general may benefit from starting prophylactic-dose heparin therapy early. One randomized trial found that starting heparin in a low subcutaneous dose the day after an ICH decreased the risk of thromboembolism without increasing the risk of rebleeding.49 Another study also found no increased risk of rebleeding with early prophylactic-dose subcutaneous heparin.50
As the benefit appears to outweigh the risk, national guidelines suggest starting subcutaneous heparin early in all ICH patients, including those not previously on warfarin.42,43
Commonly used heparinoid regimens include unfractionated heparin 5,000 units subcutaneously twice a day; enoxaparin (Lovenox) 40 mg once a day; and dalteparin (Fragmin) 5,000 units once a day.51 In addition, all patients should receive optimal mechanical thromboprophylaxis, including graduated compression stockings or intermittent pneumatic compression stockings, or both.
LONG-TERM MANAGEMENT: ICH RECURRENCE VS THROMBOEMBOLISM
Risk of ICH recurrence on warfarin is not precisely known
Overall, the risk of ICH recurrence is about 1% at 3 months, and warfarin likely increases this risk.42,52 Unfortunately, the risk of ICH recurrence in patients on anticoagulation therapy after a first ICH is not clear, and no population-based study has clarified this risk. Therefore, the best we can do at present is to try to estimate the risk of recurrent warfarin-related ICH by separately examining two issues:
The risk of ICH recurrence in general
The risk of major bleeding (including ICH) in the general population of patients on warfarin.
The risk of ICH recurrence in general is about 2% to 4% per patient-year.52–54 However, this risk appears to be a function of the underlying vasculopathy. ICH location is often used as a surrogate for underlying cause. Most ICHs in deep hemispheric (basal ganglia, thalamus) or brainstem territories are likely caused by hypertensive vasculopathy, whereas lobar ICH is often associated with cerebral amyloid angiopathy.52–54 Presumably because of this distinction, ICH in a deep location recurs in about 2% of cases per year, compared with 4% for lobar ICH.53 The presence and number of microbleeds on T2-weighted gradient-echo magnetic resonance imaging appear to predict ICH recurrence; microbleeds likely are markers of more severe or widespread underlying vasculopathy.55–57
A genetic risk factor for the recurrence of lobar ICH is apolipoprotein E genotype58; future studies may highlight genetic variations that specifically modify the risk of warfarin-related ICH.59 Unfortunately, there is currently no way to modify the risk of ICH associated with cerebral amyloid angiopathy. On the other hand, in patients with hypertensive hemorrhage, antihypertensive therapy likely reduces the risk of recurrent ICH. One randomized controlled trial showed that such therapy decreased the risk of ICH by more than half.60
The risk of major bleeding in the general population of patients on warfarin may be 2% to 3% per year and is likely higher in the first month.11 The risk is higher in older patients and if the INR rises above 4.0.11,17 For some patients, it is possible to estimate the likelihood of major bleeding using validated decision-support tools that include factors such as age, sex, and medical history.11,61–64
Given the lack of data specifically addressing the risk of ICH recurrence on warfarin, the clinician is left to try to extrapolate this risk from available data using specific patient characteristics that modify the presumed risk. For example, one can combine factors such as ICH location (or better yet, the underlying cause) with decision-support tools that predict the risk of major bleeding. Close control of both blood pressure and the INR appears especially critical for patients receiving anticoagulation after ICH.11,60,65 Still, the risk does not disappear with good INR control, and most patients with anticoagulation-related ICH present with INRs within the therapeutic range.5,10,65
Long-term risk of thromboembolism depends on underlying condition
In the long term, the risk of thromboembolism depends on the reason for which the patient was originally given anticoagulation. In addition, many patients with ICH suffer decreased mobility and are therefore at higher risk of venous thromboembolism than before their event.
Nonvalvular atrial fibrillation is the most common indication for anticoagulation. For these patients, the risk of ischemic stroke is 2% to 5% per year.66,67 The system usually used to stratify this risk is CHADS2, an acronym for five key risk factors:
Congestive heart failure (1 point)
Hypertension (1 point)
Age over 75 (1 point)
Diabetes mellitus (1 point)
Prior stroke or transient ischemic attack (2 points).
The annual risk of stroke ranges from 1.9% (score of 0) to 18.2% (score of 6).68,69 In patients with nonvalvular atrial fibrillation, the excess risk of ischemic stroke without anticoagulation must be weighed against the risk of ICH recurrence.
A mechanical heart valve, another common indication, carries a risk of ischemic stroke of about 4% per year.70 A mechanical valve is traditionally considered an absolute indication for anticoagulation. However, patients with lobar ICH face a risk of recurrence that is greater than 4% per year, and so the risks of resuming anticoagulation may well outweigh the benefits.
Heart failure may be associated with a risk of ischemic stroke of 1% to 4% per year, and this is likely a function of disease severity.71
Venous thromboembolism. The risk of recurrent venous thromboembolism in patients with deep venous thrombosis or pulmonary embolism is around 4% per year.72 Given that ICH itself confers a 2% to 3% risk of these conditions, the rate of recurrence of deep venous thrombosis may well be much higher in those ICH patients who have also already had deep venous thrombosis.
Data on resuming oral anticoagulation after ICH
Several studies have examined the outcomes when oral anticoagulants were resumed after ICH (Table 2),73–76 but experts differ on when these drugs should be resumed (eg, between 1 and 10 days after onset), or even whether they should be resumed at all.19
Notably, an analysis of 52 patients found a high risk of ICH recurrence (and gastrointestinal bleeding) in patients who restarted warfarin, and a high risk of myocardial infarction and ischemic stroke in those who did not restart, with neither strategy demonstrating a clear benefit in the rate of death or disability.73 All patients with a thromboembolic event were being treated for a previous event, suggesting that secondary prevention is a stronger indication for anticoagulation than primary prevention in this population.73
IF AND WHEN TO RESTART
Two major questions to consider are whether the benefits of restarting anticoagulation outweigh the risk, and if so, when and how should anticoagulation be restarted?
Whether to restart anticoagulation
As for the risk-benefit ratio, many think that anticoagulation should be restarted only with extreme caution and possibly only in those with deep ICH or a documented history of thromboembolism.19
In one decision analysis examining whether to restart anticoagulation after ICH in patients with atrial fibrillation, the risk of thromboembolism would need to exceed 7% per year to justify restarting anticoagulation after deep ICH,67 and no risk level was high enough to justify restarting anticoagulation after lobar ICH.
For patients at sufficiently high risk of ICH recurrence, antiplatelet treatment is probably safer, as antiplatelet agents carry a substantially lower risk of bleeding.54,77–79 The American Heart Association comments that for nonvalvular atrial fibrillation, long-term anticoagulation should be avoided after spontaneous lobar ICH, but that antiplatelet agents may be considered. 42 They note that anticoagulation after nonlobar ICH may be considered depending on the indication.42
The decision to restart anticoagulation may also be a function of whether the underlying risk factor is a temporary one. For example, atrial fibrillation or a mechanical heart valve confers a long-term, ongoing risk of arterial thromboembolism, and such patients would not normally be considered for a short course of warfarin therapy. However, isolated deep vein thrombosis may only require anticoagulation for a limited time, such as 3 to 6 months.80 Perhaps for such patients the long-term outcome is maximized with a narrowly defined, temporary course of anticoagulation.
When to restart anticoagulation
As for when to restart, it is not certain how long after symptom onset the risk of ongoing bleeding continues. Clearly, the risk is high on the first day, but small after the first few days.
The European Stroke Initiative recommends that patients with a strong indication for anticoagulation, such as a history of embolic stroke with atrial fibrillation, should be restarted on warfarin after 10 to 14 days, depending on the risk of thromboembolism and ICH recurrence.43
The American Heart Association suggests that, in patients with a very high risk of thromboembolism for whom restarting warfarin is considered, warfarin may be restarted 7 to 10 days after ICH onset.42
The American College of Chest Physicians recommends starting prophylactic-dose heparin the day after an ICH, with no clear guidance on restarting warfarin.81
ALTERNATIVES TO WARFARIN
Alternatives to warfarin that show promise in reducing bleeding risk include factor Xa and direct thrombin inhibitors, which may reduce the risk of thromboembolism to an extent similar to that of warfarin, but with fewer bleeding complications.82
In patients with atrial fibrillation, the direct thrombin inhibitor dabigatran (Pradaxa) was shown to prevent ischemic stroke to a similar or greater degree than warfarin, with fewer bleeding complications.83 Further patient follow-up is under way to ensure that this drug does not cause liver problems, as did a similarly designed predecessor.84
The availability of this and other agents in various stages of development82 will probably not make warfarin extinct. Rather, they may change the “tipping point,” the threshold at which the risk of thromboembolism is high enough to justify the risks associated with restarting warfarin therapy. In addition, clinical decision tools clarifying the individual patient’s risk of thromboembolism vs the risk of ICH recurrence will help physicians tailor the therapy to the patient.
For the moment, in situations in which the decision is difficult, maximizing the use of antiplatelet agents offers the best hope.85
RECOMMENDATIONS IN LIEU OF GUIDELINES
No guideline can broadly cover every clinical scenario. Many factors go into assessing a patient’s risk of hematoma expansion or recurrent hemorrhage (Table 3) and the extent to which anticoagulation can reduce the risk of thromboembolism.
In the short term, most patients with ICH will likely benefit from acute reversal of anticoagulation, followed by gradual reinstitution of prophylactic-dose anticoagulation after the first 24 to 72 hours.
In the long term, many patients with lobar hemorrhage, cerebral amyloid angiopathy, or other risk factors may remain at higher risk of anticoagulant-related ICH recurrence than of fatal or disabling thromboembolic events and would therefore be best managed without anticoagulants. Conversely, those with deep hemispheric ICH, hypertension that can be well controlled, and a high risk of disabling thromboembolism may receive a net benefit from restarting anticoagulation.
Wysowski DK, Nourjah P, Swartz L. Bleeding complications with warfarin use: a prevalent adverse effect resulting in regulatory action. Arch Intern Med2007; 167:1414–1419.
Choudhry NK, Anderson GM, Laupacis A, Ross-Degnan D, Normand SL, Soumerai SB. Impact of adverse events on prescribing warfarin in patients with atrial fibrillation: matched pair analysis. BMJ2006; 332:141–145.
Choudhry NK, Soumerai SB, Normand SL, Ross-Degnan D, Laupacis A, Anderson GM. Warfarin prescribing in atrial fibrillation: the impact of physician, patient, and hospital characteristics. Am J Med2006; 119:607–615.
Fang MC, Go AS, Chang Y, et al. Death and disability from warfarin-associated intracranial and extracranial hemorrhages. Am J Med2007; 120:700–705.
Rosand J, Eckman MH, Knudsen KA, Singer DE, Greenberg SM. The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage. Arch Intern Med2004; 164:880–884.
Dixon AA, Holness RO, Howes WJ, Garner JB. Spontaneous intracerebral haemorrhage: an analysis of factors affecting prognosis. Can J Neurol Sci1985; 12:267–271.
Broderick JP, Adams HP, Barsan W, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a statement for healthcare professionals from a special writing group of the Stroke Council, American Heart Association. Stroke1999; 30:905–915.
Flibotte JJ, Hagan N, O’Donnell J, Greenberg SM, Rosand J. Warfarin, hematoma expansion, and outcome of intracerebral hemorrhage. Neurology2004; 63:1059–1064.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):257S–298S.
Flaherty ML, Kissela B, Woo D, et al. The increasing incidence of anticoagulant-associated intracerebral hemorrhage. Neurology2007; 68:116–121.
Lee SB, Manno EM, Layton KF, Wijdicks EF. Progression of warfarin-associated intracerebral hemorrhage after INR normalization with FFP. Neurology2006; 67:1272–1274.
Huttner HB, Schellinger PD, Hartmann M, et al. Hematoma growth and outcome in treated neurocritical care patients with intracerebral hemorrhage related to oral anticoagulant therapy: comparison of acute treatment strategies using vitamin K, fresh frozen plasma, and prothrombin complex concentrates. Stroke2006; 37:1465–1470.
Sjöblom L, Hårdemark HG, Lindgren A, et al. Management and prognostic features of intracerebral hemorrhage during anticoagulant therapy: a Swedish multicenter study. Stroke2001; 32:2567–2574.
Yasaka M, Minematsu K, Naritomi H, Sakata T, Yamaguchi T. Predisposing factors for enlargement of intracerebral hemorrhage in patients treated with warfarin. Thromb Haemost2003; 89:278–283.
Hylek EM, Evans-Molina C, Shea C, Henault LE, Regan S. Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation. Circulation2007; 115:2689–2696.
Goldstein JN, Thomas SH, Frontiero V, et al. Timing of fresh frozen plasma administration and rapid correction of coagulopathy in warfarin-related intracerebral hemorrhage. Stroke2006; 37:151–155.
Aguilar MI, Hart RG, Kase CS, et al. Treatment of warfarin-associated intracerebral hemorrhage: literature review and expert opinion. Mayo Clin Proc2007; 82:82–92.
Guidelines on oral anticoagulation: third edition. Br J Haematol1998; 101:374–387.
Baglin TP, Keeling DM, Watson HG; British Committee for Standards in Haematology. Guidelines on oral anticoagulation (warfarin): third edition—2005 update. Br J Haematol2006; 132:277–285.
O’Shaughnessy DF, Atterbury C, Bolton Maggs P, et al; British Committee for Standards in Haematology, Blood Transfusion Task Force. Guidelines for the use of fresh-frozen plasma, cryoprecipitate and cryosupernatant. Br J Haematol2004; 126:11–28.
Baker RI, Coughlin PB, Gallus AS, Harper PL, Salem HH, Wood EM; Warfarin Reversal Consensus Group. Warfarin reversal: consensus guidelines, on behalf of the Australasian Society of Thrombosis and Haemostasis. Med J Aust2004; 181:492–497.
Davis SM, Broderick J, Hennerici M, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology2006; 66:1175–1181.
Broderick JP, Diringer MN, Hill MD, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Determinants of intracerebral hemorrhage growth: an exploratory analysis. Stroke2007; 38:1072–1075.
Goldstein JN, Fazen LE, Snider R, et al. Contrast extravasation on CT angiography predicts hematoma expansion in intracerebral hemorrhage. Neurology2007; 68:889–894.
Kazui S, Naritomi H, Yamamoto H, Sawada T, Yamaguchi T. Enlargement of spontaneous intracerebral hemorrhage. Incidence and time course. Stroke1996; 27:1783–1787.
Kim J, Smith A, Hemphill JC, et al. Contrast extravasation on CT predicts mortality in primary intracerebral hemorrhage. AJNR Am J Neuroradiol2008; 29:520–525.
Wada R, Aviv RI, Fox AJ, et al. CT angiography “spot sign” predicts hematoma expansion in acute intracerebral hemorrhage. Stroke2007; 38:1257–1262.
Barras CD, Tress BM, Christensen S, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Density and shape as CT predictors of intracerebral hemorrhage growth. Stroke2009; 40:1325–1331.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. Systematic characterization of the computed tomography angiography spot sign in primary intracerebral hemorrhage identifies patients at highest risk for hematoma expansion: the spot sign score. Stroke2009; 40:2994–3000.
Fujii Y, Takeuchi S, Sasaki O, Minakawa T, Tanaka R. Multivariate analysis of predictors of hematoma enlargement in spontaneous intracerebral hemorrhage. Stroke1998; 29:1160–1166.
Ederies A, Demchuk A, Chia T, et al. Postcontrast CT extravasation is associated with hematoma expansion in CTA spot negative patients. Stroke2009; 40:1672–1676.
Delgado P, Alvarez-Sabín J, Abilleira S, et al. Plasma d-dimer predicts poor outcome after acute intracerebral hemorrhage. Neurology2006; 67:94–98.
Naidech AM, Jovanovic B, Liebling S, et al. Reduced platelet activity is associated with early clot growth and worse 3-month outcome after intracerebral hemorrhage. Stroke2009; 40:2398–2401.
Silva Y, Leira R, Tejada J, Lainez JM, Castillo J, Dávalos A; Stroke Project, Cerebrovascular Diseases Group of the Spanish Neurological Society. Molecular signatures of vascular injury are associated with early growth of intracerebral hemorrhage. Stroke2005; 36:86–91.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Moussouttas M, Malhotra R, Fernandez L, et al. Role of antiplatelet agents in hematoma expansion during the acute period of intracerebral hemorrhage. Neurocrit Care2010; 12:24–29.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. The spot sign score in primary intracerebral hemorrhage identifies patients at highest risk of in-hospital mortality and poor outcome among survivors. Stroke2010; 41:54–60.
Broderick J, Connolly S, Feldmann E, et al; American Heart Association; American Stroke Association Stroke Council; High Blood Pressure Research Council; Quality of Care and Outcomes in Research Interdisciplinary Working Group. Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Stroke2007; 38:2001–2023.
Steiner T, Kaste M, Forsting M, et al. Recommendations for the management of intracranial haemorrhage - part I: spontaneous intracerebral haemorrhage. The European Stroke Initiative Writing Committee and the Writing Committee for the EUSI Executive Committee. Cerebrovasc Dis2006; 22:294–316.
Steiner T, Rosand J, Diringer M. Intracerebral hemorrhage associated with oral anticoagulant therapy: current practices and unresolved questions. Stroke2006; 37:256–262.
Goldstein JN, Fazen LE, Wendell L, et al. Risk of thromboembolism following acute intracerebral hemorrhage. Neurocrit Care2009; 10:28–34.
Christensen MC, Dawson J, Vincent C. Risk of thromboembolic complications after intracerebral hemorrhage according to ethnicity. Adv Ther2008; 25:831–841.
Lacut K, Bressollette L, Le Gal G, et al; VICTORIAh (Venous Intermittent Compression and Thrombosis Occurrence Related to Intracerebral Acute hemorrhage) Investigators. Prevention of venous thrombosis in patients with acute intracerebral hemorrhage. Neurology2005; 65:865–869.
Diringer MN, Skolnick BE, Mayer SA, et al. Thromboembolic events with recombinant activated factor VII in spontaneous intracerebral hemorrhage: results from the Factor Seven for Acute Hemorrhagic Stroke (FAST) trial. Stroke2010; 41:48–53.
Boeer A, Voth E, Henze T, Prange HW. Early heparin therapy in patients with spontaneous intracerebral haemorrhage. J Neurol Neurosurg Psychiatry1991; 54:466–467.
Dickmann U, Voth E, Schicha H, Henze T, Prange H, Emrich D. Heparin therapy, deep-vein thrombosis and pulmonary embolism after intracerebral hemorrhage. Klin Wochenschr1988; 66:1182–1183.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Vermeer SE, Algra A, Franke CL, Koudstaal PJ, Rinkel GJ. Long-term prognosis after recovery from primary intracerebral hemorrhage. Neurology2002; 59:205–209.
Bailey RD, Hart RG, Benavente O, Pearce LA. Recurrent brain hemorrhage is more frequent than ischemic stroke after intracranial hemorrhage. Neurology2001; 56:773–777.
Viswanathan A, Rakich SM, Engel C, et al. Antiplatelet use after intracerebral hemorrhage. Neurology2006; 66:206–209.
Greenberg SM, Eng JA, Ning M, Smith EE, Rosand J. Hemorrhage burden predicts recurrent intracerebral hemorrhage after lobar hemorrhage. Stroke2004; 35:1415–1420.
Lee SH, Ryu WS, Roh JK. Cerebral microbleeds are a risk factor for warfarin-related intracerebral hemorrhage. Neurology2009; 72:171–176.
Ueno H, Naka H, Ohshita T, et al. Association between cerebral microbleeds on T2*-weighted MR images and recurrent hemorrhagic stroke in patients treated with warfarin following ischemic stroke. AJNR Am J Neuroradiol2008; 29:1483–1486.
O’Donnell HC, Rosand J, Knudsen KA, et al. Apolipoprotein E genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med2000; 342:240–245.
Genes for Cerebral Hemorrhage on Anticoagulation (GOCHA) Collaborative Group. Exploiting common genetic variation to make anticoagulation safer. Stroke2009; 40(suppl 3):S64–S66.
Tzourio C, Arima H, Harrap S, et al. APOE genotype, ethnicity, and the risk of cerebral hemorrhage. Neurology2008; 70:1322–1328.
Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for predicting the risk of major bleeding in outpatients treated with warfarin. Am J Med1998; 105:91–99.
Kuijer PM, Hutten BA, Prins MH, Büller HR. Prediction of the risk of bleeding during anticoagulant treatment for venous thromboembolism. Arch Intern Med1999; 159:457–460.
Wess ML, Schauer DP, Johnston JA, et al. Application of a decision support tool for anticoagulation in patients with non-valvular atrial fibrillation. J Gen Intern Med2008; 23:411–417.
Singer DE, Chang Y, Fang MC, et al. Should patient characteristics influence target anticoagulation intensity for stroke prevention in nonvalvular atrial fibrillation? The ATRIA study. Circ Cardiovasc Qual Outcomes2009; 2:297–304.
Fang MC, Chang Y, Hylek EM, et al. Advanced age, anticoagulation intensity, and risk for intracranial hemorrhage among patients taking warfarin for atrial fibrillation. Ann Intern Med2004; 141:745–752.
Singer DE, Chang Y, Fang MC, et al. The net clinical benefit of warfarin anticoagulation in atrial fibrillation. Ann Intern Med2009; 151:297–305.
Eckman MH, Rosand J, Knudsen KA, Singer DE, Greenberg SM. Can patients be anticoagulated after intracerebral hemorrhage? A decision analysis. Stroke2003; 34:1710–1716.
Baruch L, Gage BF, Horrow J, et al. Can patients at elevated risk of stroke treated with anticoagulants be further risk stratified?Stroke2007; 38:2459–2463.
Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the National Registry of Atrial Fibrillation. JAMA2001; 285:2864–2870.
Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation1994; 89:635–641.
Freudenberger RS, Hellkamp AS, Halperin JL, et al; SCD-HeFT Investigators. Risk of thromboembolism in heart failure: an analysis from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation2007; 115:2637–2641.
Prandoni P, Lensing AW, Cogo A, et al. The long-term clinical course of acute deep venous thrombosis. Ann Intern Med1996; 125:1–7.
Claassen DO, Kazemi N, Zubkov AY, Wijdicks EF, Rabinstein AA. Restarting anticoagulation therapy after warfarin-associated intracerebral hemorrhage. Arch Neurol2008; 65:1313–1318.
De Vleeschouwer S, Van Calenbergh F, van Loon J, Nuttin B, Goffin J, Plets C. Risk analysis of thrombo-embolic and recurrent bleeding events in the management of intracranial haemorrhage due to oral anticoagulation. Acta Chir Belg2005; 105:268–274.
Butler AC, Tait RC. Restarting anticoagulation in prosthetic heart valve patients after intracranial haemorrhage: a 2-year follow-up. Br J Haematol1998; 103:1064–1066.
Bertram M, Bonsanto M, Hacke W, Schwab S. Managing the therapeutic dilemma: patients with spontaneous intracerebral hemorrhage and urgent need for anticoagulation. J Neurol2000; 247:209–214.
Taylor FC, Cohen H, Ebrahim S. Systematic review of long term anticoagulation or antiplatelet treatment in patients with nonrheumatic atrial fibrillation. BMJ2001; 322:321–326.
Flaherty ML, Tao H, Haverbusch M, et al. Warfarin use leads to larger intracerebral hematomas. Neurology2008; 71:1084–1089.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Snow V, Qaseem A, Barry P, et al; American College of Physicians; American Academy of Family Physicians Panel on Deep Venous Thrombosis/Pulmonary Embolism. Management of venous thromboembolism: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med2007; 146:204–210.
Albers GW, Amarenco P, Easton JD, Sacco RL, Teal PA; merican College of Chest Physicians. Antithrombotic and thrombolytic therapy for ischemic stroke: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):630S–639S.
Hankey GJ, Eikelboom JW. Antithrombotic drugs for patients with ischaemic stroke and transient ischaemic attack to prevent recurrent major vascular events. Lancet Neurol2010; 9:273–284.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Schulman S, Wåhlander K, Lundström T, Clason SB, Eriksson H; THRIVE III Investigators. Secondary prevention of venous thromboembolism with the oral direct thrombin inhibitor ximelagatran. N Engl J Med2003; 349:1713–1721.
Singer DE, Albers GW, Dalen JE, et al; American College of Chest Physicians. Antithrombotic therapy in atrial fibrillation: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):546S–592S.
Joshua N. Goldstein, MD, PhD Assistant Professor, Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA
Steven M. Greenberg, MD, PhD Director, Hemorrhagic Stroke Research Program, Department of Neurology, and the Vascular Center, Massachusetts General Hospital, Boston, MA
Address: Joshua N. Goldstein, MD, PhD, Department of Emergency Medicine, Massachusetts General Hospital, Zero Emerson Place, Suite 3B, Boston, MA 02114; e-mail [email protected]
Dr. Goldstein has acted as a consultant and a member of advisory committees or review panels for CSL Behring.
Joshua N. Goldstein, MD, PhD Assistant Professor, Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA
Steven M. Greenberg, MD, PhD Director, Hemorrhagic Stroke Research Program, Department of Neurology, and the Vascular Center, Massachusetts General Hospital, Boston, MA
Address: Joshua N. Goldstein, MD, PhD, Department of Emergency Medicine, Massachusetts General Hospital, Zero Emerson Place, Suite 3B, Boston, MA 02114; e-mail [email protected]
Dr. Goldstein has acted as a consultant and a member of advisory committees or review panels for CSL Behring.
Author and Disclosure Information
Joshua N. Goldstein, MD, PhD Assistant Professor, Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA
Steven M. Greenberg, MD, PhD Director, Hemorrhagic Stroke Research Program, Department of Neurology, and the Vascular Center, Massachusetts General Hospital, Boston, MA
Address: Joshua N. Goldstein, MD, PhD, Department of Emergency Medicine, Massachusetts General Hospital, Zero Emerson Place, Suite 3B, Boston, MA 02114; e-mail [email protected]
Dr. Goldstein has acted as a consultant and a member of advisory committees or review panels for CSL Behring.
If a patient taking warfarin (Coumadin) or other anticoagulant drug suffers an intracerebral hemorrhage (ICH) and survives, the physician faces the dilemma of whether to resume the anticoagulant. On one hand, the drug was prescribed because the patient was at risk of a thromboembolic event such as stroke or pulmonary embolism. On the other hand, warfarin use may increase the risk of another ICH.
Unfortunately, we have little evidence from clinical trials on which to base the decision. Nevertheless, we believe that in selected patients the potential benefit of resuming anticoagulation outweighs the considerable risk.
In the pages that follow, we summarize when and how anticoagulation therapy should be resumed after ICH.
A DEADLY COMPLICATION OF ANTICOAGULANT THERAPY
Intracranial bleeding is the most feared and the most deadly complication of oral anticoagulant therapy.1 The substantial risks associated with oral anticoagulants likely account for these drugs being underprescribed in patients who have indications for them.2–4
While bleeding is the major risk, not all bleeding events are equally damaging. Extracranial bleeding (eg, gastrointestinal bleeding, hematuria, epistaxis) leads to death or disability in only 3% of cases, whereas intracranial bleeding such as ICH leads to death or disability 76% of cases.5
Even without anticoagulation, ICH is the deadliest form of stroke,6–9 and if the patient has been taking warfarin, the risk of disability and death is substantially higher.6,10 Warfarin has a striking effect on the incidence and outcomes of ICH. While the overall incidence of ICH in the general population is approximately 25 per 100,000 person-years, the incidence in patients on warfarin is exponentially higher, at 2 to 3 per 100 per year, and appears to be increasing.11,12 In addition, once ICH occurs, the risk of death is up to twice as high in those on warfarin.6 The bulk of this effect is likely due to a higher risk of ongoing bleeding after the event.10,13–16
Major risk factors for ICH in patients taking oral anticoagulants include a higher international normalized ratio (INR) and older age.11,17
TWO KEY QUESTIONS
Once a patient is diagnosed with warfarin-related ICH, clinicians typically take urgent measures to restore normal coagulation, hoping to limit ongoing bleeding and improve outcome.18,19
The higher the INR at presentation, the greater the risk of death.6 In addition, in retrospective studies, some authors have noted that earlier correction of the INR is associated with better outcome.14,16
While emergency reversal of warfarin is widely considered standard treatment in the acute phase,20–24 concern persists about its safety in patients at high risk of thromboembolism.
Until the results of clinical trials are available, decisions about whether to reverse and when to resume anticoagulation hinge on two questions:
In the acute phase, how does the risk of further bleeding (hematoma expansion) compare with the short-term risk of thromboembolism?
In the chronic phase, how does the risk of recurrent hemorrhage compare with the excess risk of thromboembolism if the patient does not resume anticoagulation therapy?
ACUTELY, THE RISK OF BLEEDING OUTWEIGHS THAT OF CLOTTING
High risk of hematoma expansion after ICH
Unfortunately, continued bleeding is common after ICH. In patients who present within 3 hours of symptom onset, 26% of hematomas expand more than 33% over the first hour, and another 12% expand this amount over the next 20 hours.6 In warfarin-associated ICH, up to 50% of patients develop this level of hematoma expansion, but it appears to take place over a more prolonged period of time.10,13–16 Over 70% of patients presenting acutely develop at least some amount of expansion within 24 hours.25 Therefore, the risk of hematoma expansion in the first 24 hours is likely so high that patients cannot safely receive anticoagulants during this time frame.
But not all patients are at equal risk of hematoma expansion. Several features are associated with higher risk (Table 1)10,26–40:
A large hematoma volume on presentation is a significant predictor of expansion, possibly reflecting a more severe underlying insult.26,27
Early presentation, especially within 3 hours of symptom onset, also appears to mark those at higher risk, presumably because such patients undergo computed tomography (CT) while still bleeding.26,27
Figure 1. The “spot sign” (arrow), contrast extravasation after contrast-enhanced computed tomography, is associated with a high risk of hematoma expansion.For those on warfarin, a higher INR is a significant predictor, not just of higher risk, but also of a more delayed expansion.10,28
Certain radiographic findings indicate higher risk. One is the “spot sign,” ie, contrast extravasation after contrast-enhanced CT27,29–31 (Figure 1). Apparently, the more spots present, and the denser the contrast, the greater the risk, an observation that has led to a proposed “spot-sign score” that may predict both expansion and poor outcome. 32,41
Given the high risk of hematoma expansion in the early phase, and given our inability to predict hematoma expansion, most authorities recommend immediate reversal of anticoagulation after diagnosis.42–44 Reversal of anticoagulation typically includes intravenous vitamin K, which begins to act within several hours, and repletion of coagulation factors, which act within minutes (prothrombin complex concentrates and recombinant factor VIIa [NovoSeven]) or a few hours (fresh frozen plasma).1
Dosages:
Vitamin K 5 to 10 mg intravenously
Prothrombin complex concentrates 10 to 50 U/kg
Recombinant factor VIIa 40 to 80 μg/kg
Fresh frozen plasma 10 to 50 U/kg.
Risk of thromboembolism after ICH: Ongoing and cumulative
Thromboembolism after ICH is a major concern, for two main reasons.
First, patients on oral anticoagulation typically have a preexisting risk factor and are thus at higher risk of a thromboembolic event, particularly while they are off anticoagulation. Patients with atrial fibrillation or a mechanical valve are at risk of arterial events such as ischemic stroke, whereas patients with a known venous thromboembolic condition such as deep venous thrombosis or pulmonary embolism are at risk of extension of the thrombosis or recurrence of a venous thrombotic event.
Second, ICH itself increases the risk of arterial and venous thromboembolic events. Including patients not previously on anticoagulation, this risk is as high as 7% during the initial hospitalization and 9% during the first 90 days.45,46 Worth noting is that patients who previously received anticoagulant drugs (and who are off this therapy in the acute phase) are at no higher risk of thromboembolism compared with those who never received anticoagulants.45
However, while the risk of hematoma expansion is highest at presentation and then decreases with time, the risk of thromboembolism (particularly venous thromboembolism) is ongoing and cumulative. Arterial thromboembolism is more likely to occur early, within the first week, whereas venous thromboembolism can occur later.45
Overall, studies have estimated the short-term risk of pulmonary embolism to be 1% to 2%, deep venous thrombosis 1% to 4%, myocardial ischemia about 2%, and cerebral ischemia 2% to 3%.45,46 However, when patients are actively screened, the incidence of asymptomatic deep venous thrombosis is found to be as high as 16% in the first 10 days,47 and evidence of myocardial ischemia can be detected in up to 27% of patients.48
Therefore, the risk of hematoma expansion appears to be high and the risk of thromboembolism appears to be low during the first day after ICH. Over the next days, as the risk of hematoma expansion recedes, this ratio shifts.
Studies of in-hospital anticoagulation after ICH
The data on restarting oral anticoagulation in the acute phase are sparse. In practice, clinicians typically start heparinoids in low subcutaneous doses to prevent deep venous thrombosis and, after the first few days or a week, consider increasing to a full anticoagulation dose or starting an oral anticoagulant and subsequently discontinuing the heparin when the INR is in the therapeutic range (see discussion below).
ICH patients in general may benefit from starting prophylactic-dose heparin therapy early. One randomized trial found that starting heparin in a low subcutaneous dose the day after an ICH decreased the risk of thromboembolism without increasing the risk of rebleeding.49 Another study also found no increased risk of rebleeding with early prophylactic-dose subcutaneous heparin.50
As the benefit appears to outweigh the risk, national guidelines suggest starting subcutaneous heparin early in all ICH patients, including those not previously on warfarin.42,43
Commonly used heparinoid regimens include unfractionated heparin 5,000 units subcutaneously twice a day; enoxaparin (Lovenox) 40 mg once a day; and dalteparin (Fragmin) 5,000 units once a day.51 In addition, all patients should receive optimal mechanical thromboprophylaxis, including graduated compression stockings or intermittent pneumatic compression stockings, or both.
LONG-TERM MANAGEMENT: ICH RECURRENCE VS THROMBOEMBOLISM
Risk of ICH recurrence on warfarin is not precisely known
Overall, the risk of ICH recurrence is about 1% at 3 months, and warfarin likely increases this risk.42,52 Unfortunately, the risk of ICH recurrence in patients on anticoagulation therapy after a first ICH is not clear, and no population-based study has clarified this risk. Therefore, the best we can do at present is to try to estimate the risk of recurrent warfarin-related ICH by separately examining two issues:
The risk of ICH recurrence in general
The risk of major bleeding (including ICH) in the general population of patients on warfarin.
The risk of ICH recurrence in general is about 2% to 4% per patient-year.52–54 However, this risk appears to be a function of the underlying vasculopathy. ICH location is often used as a surrogate for underlying cause. Most ICHs in deep hemispheric (basal ganglia, thalamus) or brainstem territories are likely caused by hypertensive vasculopathy, whereas lobar ICH is often associated with cerebral amyloid angiopathy.52–54 Presumably because of this distinction, ICH in a deep location recurs in about 2% of cases per year, compared with 4% for lobar ICH.53 The presence and number of microbleeds on T2-weighted gradient-echo magnetic resonance imaging appear to predict ICH recurrence; microbleeds likely are markers of more severe or widespread underlying vasculopathy.55–57
A genetic risk factor for the recurrence of lobar ICH is apolipoprotein E genotype58; future studies may highlight genetic variations that specifically modify the risk of warfarin-related ICH.59 Unfortunately, there is currently no way to modify the risk of ICH associated with cerebral amyloid angiopathy. On the other hand, in patients with hypertensive hemorrhage, antihypertensive therapy likely reduces the risk of recurrent ICH. One randomized controlled trial showed that such therapy decreased the risk of ICH by more than half.60
The risk of major bleeding in the general population of patients on warfarin may be 2% to 3% per year and is likely higher in the first month.11 The risk is higher in older patients and if the INR rises above 4.0.11,17 For some patients, it is possible to estimate the likelihood of major bleeding using validated decision-support tools that include factors such as age, sex, and medical history.11,61–64
Given the lack of data specifically addressing the risk of ICH recurrence on warfarin, the clinician is left to try to extrapolate this risk from available data using specific patient characteristics that modify the presumed risk. For example, one can combine factors such as ICH location (or better yet, the underlying cause) with decision-support tools that predict the risk of major bleeding. Close control of both blood pressure and the INR appears especially critical for patients receiving anticoagulation after ICH.11,60,65 Still, the risk does not disappear with good INR control, and most patients with anticoagulation-related ICH present with INRs within the therapeutic range.5,10,65
Long-term risk of thromboembolism depends on underlying condition
In the long term, the risk of thromboembolism depends on the reason for which the patient was originally given anticoagulation. In addition, many patients with ICH suffer decreased mobility and are therefore at higher risk of venous thromboembolism than before their event.
Nonvalvular atrial fibrillation is the most common indication for anticoagulation. For these patients, the risk of ischemic stroke is 2% to 5% per year.66,67 The system usually used to stratify this risk is CHADS2, an acronym for five key risk factors:
Congestive heart failure (1 point)
Hypertension (1 point)
Age over 75 (1 point)
Diabetes mellitus (1 point)
Prior stroke or transient ischemic attack (2 points).
The annual risk of stroke ranges from 1.9% (score of 0) to 18.2% (score of 6).68,69 In patients with nonvalvular atrial fibrillation, the excess risk of ischemic stroke without anticoagulation must be weighed against the risk of ICH recurrence.
A mechanical heart valve, another common indication, carries a risk of ischemic stroke of about 4% per year.70 A mechanical valve is traditionally considered an absolute indication for anticoagulation. However, patients with lobar ICH face a risk of recurrence that is greater than 4% per year, and so the risks of resuming anticoagulation may well outweigh the benefits.
Heart failure may be associated with a risk of ischemic stroke of 1% to 4% per year, and this is likely a function of disease severity.71
Venous thromboembolism. The risk of recurrent venous thromboembolism in patients with deep venous thrombosis or pulmonary embolism is around 4% per year.72 Given that ICH itself confers a 2% to 3% risk of these conditions, the rate of recurrence of deep venous thrombosis may well be much higher in those ICH patients who have also already had deep venous thrombosis.
Data on resuming oral anticoagulation after ICH
Several studies have examined the outcomes when oral anticoagulants were resumed after ICH (Table 2),73–76 but experts differ on when these drugs should be resumed (eg, between 1 and 10 days after onset), or even whether they should be resumed at all.19
Notably, an analysis of 52 patients found a high risk of ICH recurrence (and gastrointestinal bleeding) in patients who restarted warfarin, and a high risk of myocardial infarction and ischemic stroke in those who did not restart, with neither strategy demonstrating a clear benefit in the rate of death or disability.73 All patients with a thromboembolic event were being treated for a previous event, suggesting that secondary prevention is a stronger indication for anticoagulation than primary prevention in this population.73
IF AND WHEN TO RESTART
Two major questions to consider are whether the benefits of restarting anticoagulation outweigh the risk, and if so, when and how should anticoagulation be restarted?
Whether to restart anticoagulation
As for the risk-benefit ratio, many think that anticoagulation should be restarted only with extreme caution and possibly only in those with deep ICH or a documented history of thromboembolism.19
In one decision analysis examining whether to restart anticoagulation after ICH in patients with atrial fibrillation, the risk of thromboembolism would need to exceed 7% per year to justify restarting anticoagulation after deep ICH,67 and no risk level was high enough to justify restarting anticoagulation after lobar ICH.
For patients at sufficiently high risk of ICH recurrence, antiplatelet treatment is probably safer, as antiplatelet agents carry a substantially lower risk of bleeding.54,77–79 The American Heart Association comments that for nonvalvular atrial fibrillation, long-term anticoagulation should be avoided after spontaneous lobar ICH, but that antiplatelet agents may be considered. 42 They note that anticoagulation after nonlobar ICH may be considered depending on the indication.42
The decision to restart anticoagulation may also be a function of whether the underlying risk factor is a temporary one. For example, atrial fibrillation or a mechanical heart valve confers a long-term, ongoing risk of arterial thromboembolism, and such patients would not normally be considered for a short course of warfarin therapy. However, isolated deep vein thrombosis may only require anticoagulation for a limited time, such as 3 to 6 months.80 Perhaps for such patients the long-term outcome is maximized with a narrowly defined, temporary course of anticoagulation.
When to restart anticoagulation
As for when to restart, it is not certain how long after symptom onset the risk of ongoing bleeding continues. Clearly, the risk is high on the first day, but small after the first few days.
The European Stroke Initiative recommends that patients with a strong indication for anticoagulation, such as a history of embolic stroke with atrial fibrillation, should be restarted on warfarin after 10 to 14 days, depending on the risk of thromboembolism and ICH recurrence.43
The American Heart Association suggests that, in patients with a very high risk of thromboembolism for whom restarting warfarin is considered, warfarin may be restarted 7 to 10 days after ICH onset.42
The American College of Chest Physicians recommends starting prophylactic-dose heparin the day after an ICH, with no clear guidance on restarting warfarin.81
ALTERNATIVES TO WARFARIN
Alternatives to warfarin that show promise in reducing bleeding risk include factor Xa and direct thrombin inhibitors, which may reduce the risk of thromboembolism to an extent similar to that of warfarin, but with fewer bleeding complications.82
In patients with atrial fibrillation, the direct thrombin inhibitor dabigatran (Pradaxa) was shown to prevent ischemic stroke to a similar or greater degree than warfarin, with fewer bleeding complications.83 Further patient follow-up is under way to ensure that this drug does not cause liver problems, as did a similarly designed predecessor.84
The availability of this and other agents in various stages of development82 will probably not make warfarin extinct. Rather, they may change the “tipping point,” the threshold at which the risk of thromboembolism is high enough to justify the risks associated with restarting warfarin therapy. In addition, clinical decision tools clarifying the individual patient’s risk of thromboembolism vs the risk of ICH recurrence will help physicians tailor the therapy to the patient.
For the moment, in situations in which the decision is difficult, maximizing the use of antiplatelet agents offers the best hope.85
RECOMMENDATIONS IN LIEU OF GUIDELINES
No guideline can broadly cover every clinical scenario. Many factors go into assessing a patient’s risk of hematoma expansion or recurrent hemorrhage (Table 3) and the extent to which anticoagulation can reduce the risk of thromboembolism.
In the short term, most patients with ICH will likely benefit from acute reversal of anticoagulation, followed by gradual reinstitution of prophylactic-dose anticoagulation after the first 24 to 72 hours.
In the long term, many patients with lobar hemorrhage, cerebral amyloid angiopathy, or other risk factors may remain at higher risk of anticoagulant-related ICH recurrence than of fatal or disabling thromboembolic events and would therefore be best managed without anticoagulants. Conversely, those with deep hemispheric ICH, hypertension that can be well controlled, and a high risk of disabling thromboembolism may receive a net benefit from restarting anticoagulation.
If a patient taking warfarin (Coumadin) or other anticoagulant drug suffers an intracerebral hemorrhage (ICH) and survives, the physician faces the dilemma of whether to resume the anticoagulant. On one hand, the drug was prescribed because the patient was at risk of a thromboembolic event such as stroke or pulmonary embolism. On the other hand, warfarin use may increase the risk of another ICH.
Unfortunately, we have little evidence from clinical trials on which to base the decision. Nevertheless, we believe that in selected patients the potential benefit of resuming anticoagulation outweighs the considerable risk.
In the pages that follow, we summarize when and how anticoagulation therapy should be resumed after ICH.
A DEADLY COMPLICATION OF ANTICOAGULANT THERAPY
Intracranial bleeding is the most feared and the most deadly complication of oral anticoagulant therapy.1 The substantial risks associated with oral anticoagulants likely account for these drugs being underprescribed in patients who have indications for them.2–4
While bleeding is the major risk, not all bleeding events are equally damaging. Extracranial bleeding (eg, gastrointestinal bleeding, hematuria, epistaxis) leads to death or disability in only 3% of cases, whereas intracranial bleeding such as ICH leads to death or disability 76% of cases.5
Even without anticoagulation, ICH is the deadliest form of stroke,6–9 and if the patient has been taking warfarin, the risk of disability and death is substantially higher.6,10 Warfarin has a striking effect on the incidence and outcomes of ICH. While the overall incidence of ICH in the general population is approximately 25 per 100,000 person-years, the incidence in patients on warfarin is exponentially higher, at 2 to 3 per 100 per year, and appears to be increasing.11,12 In addition, once ICH occurs, the risk of death is up to twice as high in those on warfarin.6 The bulk of this effect is likely due to a higher risk of ongoing bleeding after the event.10,13–16
Major risk factors for ICH in patients taking oral anticoagulants include a higher international normalized ratio (INR) and older age.11,17
TWO KEY QUESTIONS
Once a patient is diagnosed with warfarin-related ICH, clinicians typically take urgent measures to restore normal coagulation, hoping to limit ongoing bleeding and improve outcome.18,19
The higher the INR at presentation, the greater the risk of death.6 In addition, in retrospective studies, some authors have noted that earlier correction of the INR is associated with better outcome.14,16
While emergency reversal of warfarin is widely considered standard treatment in the acute phase,20–24 concern persists about its safety in patients at high risk of thromboembolism.
Until the results of clinical trials are available, decisions about whether to reverse and when to resume anticoagulation hinge on two questions:
In the acute phase, how does the risk of further bleeding (hematoma expansion) compare with the short-term risk of thromboembolism?
In the chronic phase, how does the risk of recurrent hemorrhage compare with the excess risk of thromboembolism if the patient does not resume anticoagulation therapy?
ACUTELY, THE RISK OF BLEEDING OUTWEIGHS THAT OF CLOTTING
High risk of hematoma expansion after ICH
Unfortunately, continued bleeding is common after ICH. In patients who present within 3 hours of symptom onset, 26% of hematomas expand more than 33% over the first hour, and another 12% expand this amount over the next 20 hours.6 In warfarin-associated ICH, up to 50% of patients develop this level of hematoma expansion, but it appears to take place over a more prolonged period of time.10,13–16 Over 70% of patients presenting acutely develop at least some amount of expansion within 24 hours.25 Therefore, the risk of hematoma expansion in the first 24 hours is likely so high that patients cannot safely receive anticoagulants during this time frame.
But not all patients are at equal risk of hematoma expansion. Several features are associated with higher risk (Table 1)10,26–40:
A large hematoma volume on presentation is a significant predictor of expansion, possibly reflecting a more severe underlying insult.26,27
Early presentation, especially within 3 hours of symptom onset, also appears to mark those at higher risk, presumably because such patients undergo computed tomography (CT) while still bleeding.26,27
Figure 1. The “spot sign” (arrow), contrast extravasation after contrast-enhanced computed tomography, is associated with a high risk of hematoma expansion.For those on warfarin, a higher INR is a significant predictor, not just of higher risk, but also of a more delayed expansion.10,28
Certain radiographic findings indicate higher risk. One is the “spot sign,” ie, contrast extravasation after contrast-enhanced CT27,29–31 (Figure 1). Apparently, the more spots present, and the denser the contrast, the greater the risk, an observation that has led to a proposed “spot-sign score” that may predict both expansion and poor outcome. 32,41
Given the high risk of hematoma expansion in the early phase, and given our inability to predict hematoma expansion, most authorities recommend immediate reversal of anticoagulation after diagnosis.42–44 Reversal of anticoagulation typically includes intravenous vitamin K, which begins to act within several hours, and repletion of coagulation factors, which act within minutes (prothrombin complex concentrates and recombinant factor VIIa [NovoSeven]) or a few hours (fresh frozen plasma).1
Dosages:
Vitamin K 5 to 10 mg intravenously
Prothrombin complex concentrates 10 to 50 U/kg
Recombinant factor VIIa 40 to 80 μg/kg
Fresh frozen plasma 10 to 50 U/kg.
Risk of thromboembolism after ICH: Ongoing and cumulative
Thromboembolism after ICH is a major concern, for two main reasons.
First, patients on oral anticoagulation typically have a preexisting risk factor and are thus at higher risk of a thromboembolic event, particularly while they are off anticoagulation. Patients with atrial fibrillation or a mechanical valve are at risk of arterial events such as ischemic stroke, whereas patients with a known venous thromboembolic condition such as deep venous thrombosis or pulmonary embolism are at risk of extension of the thrombosis or recurrence of a venous thrombotic event.
Second, ICH itself increases the risk of arterial and venous thromboembolic events. Including patients not previously on anticoagulation, this risk is as high as 7% during the initial hospitalization and 9% during the first 90 days.45,46 Worth noting is that patients who previously received anticoagulant drugs (and who are off this therapy in the acute phase) are at no higher risk of thromboembolism compared with those who never received anticoagulants.45
However, while the risk of hematoma expansion is highest at presentation and then decreases with time, the risk of thromboembolism (particularly venous thromboembolism) is ongoing and cumulative. Arterial thromboembolism is more likely to occur early, within the first week, whereas venous thromboembolism can occur later.45
Overall, studies have estimated the short-term risk of pulmonary embolism to be 1% to 2%, deep venous thrombosis 1% to 4%, myocardial ischemia about 2%, and cerebral ischemia 2% to 3%.45,46 However, when patients are actively screened, the incidence of asymptomatic deep venous thrombosis is found to be as high as 16% in the first 10 days,47 and evidence of myocardial ischemia can be detected in up to 27% of patients.48
Therefore, the risk of hematoma expansion appears to be high and the risk of thromboembolism appears to be low during the first day after ICH. Over the next days, as the risk of hematoma expansion recedes, this ratio shifts.
Studies of in-hospital anticoagulation after ICH
The data on restarting oral anticoagulation in the acute phase are sparse. In practice, clinicians typically start heparinoids in low subcutaneous doses to prevent deep venous thrombosis and, after the first few days or a week, consider increasing to a full anticoagulation dose or starting an oral anticoagulant and subsequently discontinuing the heparin when the INR is in the therapeutic range (see discussion below).
ICH patients in general may benefit from starting prophylactic-dose heparin therapy early. One randomized trial found that starting heparin in a low subcutaneous dose the day after an ICH decreased the risk of thromboembolism without increasing the risk of rebleeding.49 Another study also found no increased risk of rebleeding with early prophylactic-dose subcutaneous heparin.50
As the benefit appears to outweigh the risk, national guidelines suggest starting subcutaneous heparin early in all ICH patients, including those not previously on warfarin.42,43
Commonly used heparinoid regimens include unfractionated heparin 5,000 units subcutaneously twice a day; enoxaparin (Lovenox) 40 mg once a day; and dalteparin (Fragmin) 5,000 units once a day.51 In addition, all patients should receive optimal mechanical thromboprophylaxis, including graduated compression stockings or intermittent pneumatic compression stockings, or both.
LONG-TERM MANAGEMENT: ICH RECURRENCE VS THROMBOEMBOLISM
Risk of ICH recurrence on warfarin is not precisely known
Overall, the risk of ICH recurrence is about 1% at 3 months, and warfarin likely increases this risk.42,52 Unfortunately, the risk of ICH recurrence in patients on anticoagulation therapy after a first ICH is not clear, and no population-based study has clarified this risk. Therefore, the best we can do at present is to try to estimate the risk of recurrent warfarin-related ICH by separately examining two issues:
The risk of ICH recurrence in general
The risk of major bleeding (including ICH) in the general population of patients on warfarin.
The risk of ICH recurrence in general is about 2% to 4% per patient-year.52–54 However, this risk appears to be a function of the underlying vasculopathy. ICH location is often used as a surrogate for underlying cause. Most ICHs in deep hemispheric (basal ganglia, thalamus) or brainstem territories are likely caused by hypertensive vasculopathy, whereas lobar ICH is often associated with cerebral amyloid angiopathy.52–54 Presumably because of this distinction, ICH in a deep location recurs in about 2% of cases per year, compared with 4% for lobar ICH.53 The presence and number of microbleeds on T2-weighted gradient-echo magnetic resonance imaging appear to predict ICH recurrence; microbleeds likely are markers of more severe or widespread underlying vasculopathy.55–57
A genetic risk factor for the recurrence of lobar ICH is apolipoprotein E genotype58; future studies may highlight genetic variations that specifically modify the risk of warfarin-related ICH.59 Unfortunately, there is currently no way to modify the risk of ICH associated with cerebral amyloid angiopathy. On the other hand, in patients with hypertensive hemorrhage, antihypertensive therapy likely reduces the risk of recurrent ICH. One randomized controlled trial showed that such therapy decreased the risk of ICH by more than half.60
The risk of major bleeding in the general population of patients on warfarin may be 2% to 3% per year and is likely higher in the first month.11 The risk is higher in older patients and if the INR rises above 4.0.11,17 For some patients, it is possible to estimate the likelihood of major bleeding using validated decision-support tools that include factors such as age, sex, and medical history.11,61–64
Given the lack of data specifically addressing the risk of ICH recurrence on warfarin, the clinician is left to try to extrapolate this risk from available data using specific patient characteristics that modify the presumed risk. For example, one can combine factors such as ICH location (or better yet, the underlying cause) with decision-support tools that predict the risk of major bleeding. Close control of both blood pressure and the INR appears especially critical for patients receiving anticoagulation after ICH.11,60,65 Still, the risk does not disappear with good INR control, and most patients with anticoagulation-related ICH present with INRs within the therapeutic range.5,10,65
Long-term risk of thromboembolism depends on underlying condition
In the long term, the risk of thromboembolism depends on the reason for which the patient was originally given anticoagulation. In addition, many patients with ICH suffer decreased mobility and are therefore at higher risk of venous thromboembolism than before their event.
Nonvalvular atrial fibrillation is the most common indication for anticoagulation. For these patients, the risk of ischemic stroke is 2% to 5% per year.66,67 The system usually used to stratify this risk is CHADS2, an acronym for five key risk factors:
Congestive heart failure (1 point)
Hypertension (1 point)
Age over 75 (1 point)
Diabetes mellitus (1 point)
Prior stroke or transient ischemic attack (2 points).
The annual risk of stroke ranges from 1.9% (score of 0) to 18.2% (score of 6).68,69 In patients with nonvalvular atrial fibrillation, the excess risk of ischemic stroke without anticoagulation must be weighed against the risk of ICH recurrence.
A mechanical heart valve, another common indication, carries a risk of ischemic stroke of about 4% per year.70 A mechanical valve is traditionally considered an absolute indication for anticoagulation. However, patients with lobar ICH face a risk of recurrence that is greater than 4% per year, and so the risks of resuming anticoagulation may well outweigh the benefits.
Heart failure may be associated with a risk of ischemic stroke of 1% to 4% per year, and this is likely a function of disease severity.71
Venous thromboembolism. The risk of recurrent venous thromboembolism in patients with deep venous thrombosis or pulmonary embolism is around 4% per year.72 Given that ICH itself confers a 2% to 3% risk of these conditions, the rate of recurrence of deep venous thrombosis may well be much higher in those ICH patients who have also already had deep venous thrombosis.
Data on resuming oral anticoagulation after ICH
Several studies have examined the outcomes when oral anticoagulants were resumed after ICH (Table 2),73–76 but experts differ on when these drugs should be resumed (eg, between 1 and 10 days after onset), or even whether they should be resumed at all.19
Notably, an analysis of 52 patients found a high risk of ICH recurrence (and gastrointestinal bleeding) in patients who restarted warfarin, and a high risk of myocardial infarction and ischemic stroke in those who did not restart, with neither strategy demonstrating a clear benefit in the rate of death or disability.73 All patients with a thromboembolic event were being treated for a previous event, suggesting that secondary prevention is a stronger indication for anticoagulation than primary prevention in this population.73
IF AND WHEN TO RESTART
Two major questions to consider are whether the benefits of restarting anticoagulation outweigh the risk, and if so, when and how should anticoagulation be restarted?
Whether to restart anticoagulation
As for the risk-benefit ratio, many think that anticoagulation should be restarted only with extreme caution and possibly only in those with deep ICH or a documented history of thromboembolism.19
In one decision analysis examining whether to restart anticoagulation after ICH in patients with atrial fibrillation, the risk of thromboembolism would need to exceed 7% per year to justify restarting anticoagulation after deep ICH,67 and no risk level was high enough to justify restarting anticoagulation after lobar ICH.
For patients at sufficiently high risk of ICH recurrence, antiplatelet treatment is probably safer, as antiplatelet agents carry a substantially lower risk of bleeding.54,77–79 The American Heart Association comments that for nonvalvular atrial fibrillation, long-term anticoagulation should be avoided after spontaneous lobar ICH, but that antiplatelet agents may be considered. 42 They note that anticoagulation after nonlobar ICH may be considered depending on the indication.42
The decision to restart anticoagulation may also be a function of whether the underlying risk factor is a temporary one. For example, atrial fibrillation or a mechanical heart valve confers a long-term, ongoing risk of arterial thromboembolism, and such patients would not normally be considered for a short course of warfarin therapy. However, isolated deep vein thrombosis may only require anticoagulation for a limited time, such as 3 to 6 months.80 Perhaps for such patients the long-term outcome is maximized with a narrowly defined, temporary course of anticoagulation.
When to restart anticoagulation
As for when to restart, it is not certain how long after symptom onset the risk of ongoing bleeding continues. Clearly, the risk is high on the first day, but small after the first few days.
The European Stroke Initiative recommends that patients with a strong indication for anticoagulation, such as a history of embolic stroke with atrial fibrillation, should be restarted on warfarin after 10 to 14 days, depending on the risk of thromboembolism and ICH recurrence.43
The American Heart Association suggests that, in patients with a very high risk of thromboembolism for whom restarting warfarin is considered, warfarin may be restarted 7 to 10 days after ICH onset.42
The American College of Chest Physicians recommends starting prophylactic-dose heparin the day after an ICH, with no clear guidance on restarting warfarin.81
ALTERNATIVES TO WARFARIN
Alternatives to warfarin that show promise in reducing bleeding risk include factor Xa and direct thrombin inhibitors, which may reduce the risk of thromboembolism to an extent similar to that of warfarin, but with fewer bleeding complications.82
In patients with atrial fibrillation, the direct thrombin inhibitor dabigatran (Pradaxa) was shown to prevent ischemic stroke to a similar or greater degree than warfarin, with fewer bleeding complications.83 Further patient follow-up is under way to ensure that this drug does not cause liver problems, as did a similarly designed predecessor.84
The availability of this and other agents in various stages of development82 will probably not make warfarin extinct. Rather, they may change the “tipping point,” the threshold at which the risk of thromboembolism is high enough to justify the risks associated with restarting warfarin therapy. In addition, clinical decision tools clarifying the individual patient’s risk of thromboembolism vs the risk of ICH recurrence will help physicians tailor the therapy to the patient.
For the moment, in situations in which the decision is difficult, maximizing the use of antiplatelet agents offers the best hope.85
RECOMMENDATIONS IN LIEU OF GUIDELINES
No guideline can broadly cover every clinical scenario. Many factors go into assessing a patient’s risk of hematoma expansion or recurrent hemorrhage (Table 3) and the extent to which anticoagulation can reduce the risk of thromboembolism.
In the short term, most patients with ICH will likely benefit from acute reversal of anticoagulation, followed by gradual reinstitution of prophylactic-dose anticoagulation after the first 24 to 72 hours.
In the long term, many patients with lobar hemorrhage, cerebral amyloid angiopathy, or other risk factors may remain at higher risk of anticoagulant-related ICH recurrence than of fatal or disabling thromboembolic events and would therefore be best managed without anticoagulants. Conversely, those with deep hemispheric ICH, hypertension that can be well controlled, and a high risk of disabling thromboembolism may receive a net benefit from restarting anticoagulation.
Wysowski DK, Nourjah P, Swartz L. Bleeding complications with warfarin use: a prevalent adverse effect resulting in regulatory action. Arch Intern Med2007; 167:1414–1419.
Choudhry NK, Anderson GM, Laupacis A, Ross-Degnan D, Normand SL, Soumerai SB. Impact of adverse events on prescribing warfarin in patients with atrial fibrillation: matched pair analysis. BMJ2006; 332:141–145.
Choudhry NK, Soumerai SB, Normand SL, Ross-Degnan D, Laupacis A, Anderson GM. Warfarin prescribing in atrial fibrillation: the impact of physician, patient, and hospital characteristics. Am J Med2006; 119:607–615.
Fang MC, Go AS, Chang Y, et al. Death and disability from warfarin-associated intracranial and extracranial hemorrhages. Am J Med2007; 120:700–705.
Rosand J, Eckman MH, Knudsen KA, Singer DE, Greenberg SM. The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage. Arch Intern Med2004; 164:880–884.
Dixon AA, Holness RO, Howes WJ, Garner JB. Spontaneous intracerebral haemorrhage: an analysis of factors affecting prognosis. Can J Neurol Sci1985; 12:267–271.
Broderick JP, Adams HP, Barsan W, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a statement for healthcare professionals from a special writing group of the Stroke Council, American Heart Association. Stroke1999; 30:905–915.
Flibotte JJ, Hagan N, O’Donnell J, Greenberg SM, Rosand J. Warfarin, hematoma expansion, and outcome of intracerebral hemorrhage. Neurology2004; 63:1059–1064.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):257S–298S.
Flaherty ML, Kissela B, Woo D, et al. The increasing incidence of anticoagulant-associated intracerebral hemorrhage. Neurology2007; 68:116–121.
Lee SB, Manno EM, Layton KF, Wijdicks EF. Progression of warfarin-associated intracerebral hemorrhage after INR normalization with FFP. Neurology2006; 67:1272–1274.
Huttner HB, Schellinger PD, Hartmann M, et al. Hematoma growth and outcome in treated neurocritical care patients with intracerebral hemorrhage related to oral anticoagulant therapy: comparison of acute treatment strategies using vitamin K, fresh frozen plasma, and prothrombin complex concentrates. Stroke2006; 37:1465–1470.
Sjöblom L, Hårdemark HG, Lindgren A, et al. Management and prognostic features of intracerebral hemorrhage during anticoagulant therapy: a Swedish multicenter study. Stroke2001; 32:2567–2574.
Yasaka M, Minematsu K, Naritomi H, Sakata T, Yamaguchi T. Predisposing factors for enlargement of intracerebral hemorrhage in patients treated with warfarin. Thromb Haemost2003; 89:278–283.
Hylek EM, Evans-Molina C, Shea C, Henault LE, Regan S. Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation. Circulation2007; 115:2689–2696.
Goldstein JN, Thomas SH, Frontiero V, et al. Timing of fresh frozen plasma administration and rapid correction of coagulopathy in warfarin-related intracerebral hemorrhage. Stroke2006; 37:151–155.
Aguilar MI, Hart RG, Kase CS, et al. Treatment of warfarin-associated intracerebral hemorrhage: literature review and expert opinion. Mayo Clin Proc2007; 82:82–92.
Guidelines on oral anticoagulation: third edition. Br J Haematol1998; 101:374–387.
Baglin TP, Keeling DM, Watson HG; British Committee for Standards in Haematology. Guidelines on oral anticoagulation (warfarin): third edition—2005 update. Br J Haematol2006; 132:277–285.
O’Shaughnessy DF, Atterbury C, Bolton Maggs P, et al; British Committee for Standards in Haematology, Blood Transfusion Task Force. Guidelines for the use of fresh-frozen plasma, cryoprecipitate and cryosupernatant. Br J Haematol2004; 126:11–28.
Baker RI, Coughlin PB, Gallus AS, Harper PL, Salem HH, Wood EM; Warfarin Reversal Consensus Group. Warfarin reversal: consensus guidelines, on behalf of the Australasian Society of Thrombosis and Haemostasis. Med J Aust2004; 181:492–497.
Davis SM, Broderick J, Hennerici M, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology2006; 66:1175–1181.
Broderick JP, Diringer MN, Hill MD, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Determinants of intracerebral hemorrhage growth: an exploratory analysis. Stroke2007; 38:1072–1075.
Goldstein JN, Fazen LE, Snider R, et al. Contrast extravasation on CT angiography predicts hematoma expansion in intracerebral hemorrhage. Neurology2007; 68:889–894.
Kazui S, Naritomi H, Yamamoto H, Sawada T, Yamaguchi T. Enlargement of spontaneous intracerebral hemorrhage. Incidence and time course. Stroke1996; 27:1783–1787.
Kim J, Smith A, Hemphill JC, et al. Contrast extravasation on CT predicts mortality in primary intracerebral hemorrhage. AJNR Am J Neuroradiol2008; 29:520–525.
Wada R, Aviv RI, Fox AJ, et al. CT angiography “spot sign” predicts hematoma expansion in acute intracerebral hemorrhage. Stroke2007; 38:1257–1262.
Barras CD, Tress BM, Christensen S, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Density and shape as CT predictors of intracerebral hemorrhage growth. Stroke2009; 40:1325–1331.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. Systematic characterization of the computed tomography angiography spot sign in primary intracerebral hemorrhage identifies patients at highest risk for hematoma expansion: the spot sign score. Stroke2009; 40:2994–3000.
Fujii Y, Takeuchi S, Sasaki O, Minakawa T, Tanaka R. Multivariate analysis of predictors of hematoma enlargement in spontaneous intracerebral hemorrhage. Stroke1998; 29:1160–1166.
Ederies A, Demchuk A, Chia T, et al. Postcontrast CT extravasation is associated with hematoma expansion in CTA spot negative patients. Stroke2009; 40:1672–1676.
Delgado P, Alvarez-Sabín J, Abilleira S, et al. Plasma d-dimer predicts poor outcome after acute intracerebral hemorrhage. Neurology2006; 67:94–98.
Naidech AM, Jovanovic B, Liebling S, et al. Reduced platelet activity is associated with early clot growth and worse 3-month outcome after intracerebral hemorrhage. Stroke2009; 40:2398–2401.
Silva Y, Leira R, Tejada J, Lainez JM, Castillo J, Dávalos A; Stroke Project, Cerebrovascular Diseases Group of the Spanish Neurological Society. Molecular signatures of vascular injury are associated with early growth of intracerebral hemorrhage. Stroke2005; 36:86–91.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Moussouttas M, Malhotra R, Fernandez L, et al. Role of antiplatelet agents in hematoma expansion during the acute period of intracerebral hemorrhage. Neurocrit Care2010; 12:24–29.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. The spot sign score in primary intracerebral hemorrhage identifies patients at highest risk of in-hospital mortality and poor outcome among survivors. Stroke2010; 41:54–60.
Broderick J, Connolly S, Feldmann E, et al; American Heart Association; American Stroke Association Stroke Council; High Blood Pressure Research Council; Quality of Care and Outcomes in Research Interdisciplinary Working Group. Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Stroke2007; 38:2001–2023.
Steiner T, Kaste M, Forsting M, et al. Recommendations for the management of intracranial haemorrhage - part I: spontaneous intracerebral haemorrhage. The European Stroke Initiative Writing Committee and the Writing Committee for the EUSI Executive Committee. Cerebrovasc Dis2006; 22:294–316.
Steiner T, Rosand J, Diringer M. Intracerebral hemorrhage associated with oral anticoagulant therapy: current practices and unresolved questions. Stroke2006; 37:256–262.
Goldstein JN, Fazen LE, Wendell L, et al. Risk of thromboembolism following acute intracerebral hemorrhage. Neurocrit Care2009; 10:28–34.
Christensen MC, Dawson J, Vincent C. Risk of thromboembolic complications after intracerebral hemorrhage according to ethnicity. Adv Ther2008; 25:831–841.
Lacut K, Bressollette L, Le Gal G, et al; VICTORIAh (Venous Intermittent Compression and Thrombosis Occurrence Related to Intracerebral Acute hemorrhage) Investigators. Prevention of venous thrombosis in patients with acute intracerebral hemorrhage. Neurology2005; 65:865–869.
Diringer MN, Skolnick BE, Mayer SA, et al. Thromboembolic events with recombinant activated factor VII in spontaneous intracerebral hemorrhage: results from the Factor Seven for Acute Hemorrhagic Stroke (FAST) trial. Stroke2010; 41:48–53.
Boeer A, Voth E, Henze T, Prange HW. Early heparin therapy in patients with spontaneous intracerebral haemorrhage. J Neurol Neurosurg Psychiatry1991; 54:466–467.
Dickmann U, Voth E, Schicha H, Henze T, Prange H, Emrich D. Heparin therapy, deep-vein thrombosis and pulmonary embolism after intracerebral hemorrhage. Klin Wochenschr1988; 66:1182–1183.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Vermeer SE, Algra A, Franke CL, Koudstaal PJ, Rinkel GJ. Long-term prognosis after recovery from primary intracerebral hemorrhage. Neurology2002; 59:205–209.
Bailey RD, Hart RG, Benavente O, Pearce LA. Recurrent brain hemorrhage is more frequent than ischemic stroke after intracranial hemorrhage. Neurology2001; 56:773–777.
Viswanathan A, Rakich SM, Engel C, et al. Antiplatelet use after intracerebral hemorrhage. Neurology2006; 66:206–209.
Greenberg SM, Eng JA, Ning M, Smith EE, Rosand J. Hemorrhage burden predicts recurrent intracerebral hemorrhage after lobar hemorrhage. Stroke2004; 35:1415–1420.
Lee SH, Ryu WS, Roh JK. Cerebral microbleeds are a risk factor for warfarin-related intracerebral hemorrhage. Neurology2009; 72:171–176.
Ueno H, Naka H, Ohshita T, et al. Association between cerebral microbleeds on T2*-weighted MR images and recurrent hemorrhagic stroke in patients treated with warfarin following ischemic stroke. AJNR Am J Neuroradiol2008; 29:1483–1486.
O’Donnell HC, Rosand J, Knudsen KA, et al. Apolipoprotein E genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med2000; 342:240–245.
Genes for Cerebral Hemorrhage on Anticoagulation (GOCHA) Collaborative Group. Exploiting common genetic variation to make anticoagulation safer. Stroke2009; 40(suppl 3):S64–S66.
Tzourio C, Arima H, Harrap S, et al. APOE genotype, ethnicity, and the risk of cerebral hemorrhage. Neurology2008; 70:1322–1328.
Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for predicting the risk of major bleeding in outpatients treated with warfarin. Am J Med1998; 105:91–99.
Kuijer PM, Hutten BA, Prins MH, Büller HR. Prediction of the risk of bleeding during anticoagulant treatment for venous thromboembolism. Arch Intern Med1999; 159:457–460.
Wess ML, Schauer DP, Johnston JA, et al. Application of a decision support tool for anticoagulation in patients with non-valvular atrial fibrillation. J Gen Intern Med2008; 23:411–417.
Singer DE, Chang Y, Fang MC, et al. Should patient characteristics influence target anticoagulation intensity for stroke prevention in nonvalvular atrial fibrillation? The ATRIA study. Circ Cardiovasc Qual Outcomes2009; 2:297–304.
Fang MC, Chang Y, Hylek EM, et al. Advanced age, anticoagulation intensity, and risk for intracranial hemorrhage among patients taking warfarin for atrial fibrillation. Ann Intern Med2004; 141:745–752.
Singer DE, Chang Y, Fang MC, et al. The net clinical benefit of warfarin anticoagulation in atrial fibrillation. Ann Intern Med2009; 151:297–305.
Eckman MH, Rosand J, Knudsen KA, Singer DE, Greenberg SM. Can patients be anticoagulated after intracerebral hemorrhage? A decision analysis. Stroke2003; 34:1710–1716.
Baruch L, Gage BF, Horrow J, et al. Can patients at elevated risk of stroke treated with anticoagulants be further risk stratified?Stroke2007; 38:2459–2463.
Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the National Registry of Atrial Fibrillation. JAMA2001; 285:2864–2870.
Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation1994; 89:635–641.
Freudenberger RS, Hellkamp AS, Halperin JL, et al; SCD-HeFT Investigators. Risk of thromboembolism in heart failure: an analysis from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation2007; 115:2637–2641.
Prandoni P, Lensing AW, Cogo A, et al. The long-term clinical course of acute deep venous thrombosis. Ann Intern Med1996; 125:1–7.
Claassen DO, Kazemi N, Zubkov AY, Wijdicks EF, Rabinstein AA. Restarting anticoagulation therapy after warfarin-associated intracerebral hemorrhage. Arch Neurol2008; 65:1313–1318.
De Vleeschouwer S, Van Calenbergh F, van Loon J, Nuttin B, Goffin J, Plets C. Risk analysis of thrombo-embolic and recurrent bleeding events in the management of intracranial haemorrhage due to oral anticoagulation. Acta Chir Belg2005; 105:268–274.
Butler AC, Tait RC. Restarting anticoagulation in prosthetic heart valve patients after intracranial haemorrhage: a 2-year follow-up. Br J Haematol1998; 103:1064–1066.
Bertram M, Bonsanto M, Hacke W, Schwab S. Managing the therapeutic dilemma: patients with spontaneous intracerebral hemorrhage and urgent need for anticoagulation. J Neurol2000; 247:209–214.
Taylor FC, Cohen H, Ebrahim S. Systematic review of long term anticoagulation or antiplatelet treatment in patients with nonrheumatic atrial fibrillation. BMJ2001; 322:321–326.
Flaherty ML, Tao H, Haverbusch M, et al. Warfarin use leads to larger intracerebral hematomas. Neurology2008; 71:1084–1089.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Snow V, Qaseem A, Barry P, et al; American College of Physicians; American Academy of Family Physicians Panel on Deep Venous Thrombosis/Pulmonary Embolism. Management of venous thromboembolism: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med2007; 146:204–210.
Albers GW, Amarenco P, Easton JD, Sacco RL, Teal PA; merican College of Chest Physicians. Antithrombotic and thrombolytic therapy for ischemic stroke: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):630S–639S.
Hankey GJ, Eikelboom JW. Antithrombotic drugs for patients with ischaemic stroke and transient ischaemic attack to prevent recurrent major vascular events. Lancet Neurol2010; 9:273–284.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Schulman S, Wåhlander K, Lundström T, Clason SB, Eriksson H; THRIVE III Investigators. Secondary prevention of venous thromboembolism with the oral direct thrombin inhibitor ximelagatran. N Engl J Med2003; 349:1713–1721.
Singer DE, Albers GW, Dalen JE, et al; American College of Chest Physicians. Antithrombotic therapy in atrial fibrillation: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):546S–592S.
Wysowski DK, Nourjah P, Swartz L. Bleeding complications with warfarin use: a prevalent adverse effect resulting in regulatory action. Arch Intern Med2007; 167:1414–1419.
Choudhry NK, Anderson GM, Laupacis A, Ross-Degnan D, Normand SL, Soumerai SB. Impact of adverse events on prescribing warfarin in patients with atrial fibrillation: matched pair analysis. BMJ2006; 332:141–145.
Choudhry NK, Soumerai SB, Normand SL, Ross-Degnan D, Laupacis A, Anderson GM. Warfarin prescribing in atrial fibrillation: the impact of physician, patient, and hospital characteristics. Am J Med2006; 119:607–615.
Fang MC, Go AS, Chang Y, et al. Death and disability from warfarin-associated intracranial and extracranial hemorrhages. Am J Med2007; 120:700–705.
Rosand J, Eckman MH, Knudsen KA, Singer DE, Greenberg SM. The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage. Arch Intern Med2004; 164:880–884.
Dixon AA, Holness RO, Howes WJ, Garner JB. Spontaneous intracerebral haemorrhage: an analysis of factors affecting prognosis. Can J Neurol Sci1985; 12:267–271.
Broderick JP, Adams HP, Barsan W, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a statement for healthcare professionals from a special writing group of the Stroke Council, American Heart Association. Stroke1999; 30:905–915.
Flibotte JJ, Hagan N, O’Donnell J, Greenberg SM, Rosand J. Warfarin, hematoma expansion, and outcome of intracerebral hemorrhage. Neurology2004; 63:1059–1064.
Schulman S, Beyth RJ, Kearon C, Levine MN; American College of Chest Physicians. Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):257S–298S.
Flaherty ML, Kissela B, Woo D, et al. The increasing incidence of anticoagulant-associated intracerebral hemorrhage. Neurology2007; 68:116–121.
Lee SB, Manno EM, Layton KF, Wijdicks EF. Progression of warfarin-associated intracerebral hemorrhage after INR normalization with FFP. Neurology2006; 67:1272–1274.
Huttner HB, Schellinger PD, Hartmann M, et al. Hematoma growth and outcome in treated neurocritical care patients with intracerebral hemorrhage related to oral anticoagulant therapy: comparison of acute treatment strategies using vitamin K, fresh frozen plasma, and prothrombin complex concentrates. Stroke2006; 37:1465–1470.
Sjöblom L, Hårdemark HG, Lindgren A, et al. Management and prognostic features of intracerebral hemorrhage during anticoagulant therapy: a Swedish multicenter study. Stroke2001; 32:2567–2574.
Yasaka M, Minematsu K, Naritomi H, Sakata T, Yamaguchi T. Predisposing factors for enlargement of intracerebral hemorrhage in patients treated with warfarin. Thromb Haemost2003; 89:278–283.
Hylek EM, Evans-Molina C, Shea C, Henault LE, Regan S. Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation. Circulation2007; 115:2689–2696.
Goldstein JN, Thomas SH, Frontiero V, et al. Timing of fresh frozen plasma administration and rapid correction of coagulopathy in warfarin-related intracerebral hemorrhage. Stroke2006; 37:151–155.
Aguilar MI, Hart RG, Kase CS, et al. Treatment of warfarin-associated intracerebral hemorrhage: literature review and expert opinion. Mayo Clin Proc2007; 82:82–92.
Guidelines on oral anticoagulation: third edition. Br J Haematol1998; 101:374–387.
Baglin TP, Keeling DM, Watson HG; British Committee for Standards in Haematology. Guidelines on oral anticoagulation (warfarin): third edition—2005 update. Br J Haematol2006; 132:277–285.
O’Shaughnessy DF, Atterbury C, Bolton Maggs P, et al; British Committee for Standards in Haematology, Blood Transfusion Task Force. Guidelines for the use of fresh-frozen plasma, cryoprecipitate and cryosupernatant. Br J Haematol2004; 126:11–28.
Baker RI, Coughlin PB, Gallus AS, Harper PL, Salem HH, Wood EM; Warfarin Reversal Consensus Group. Warfarin reversal: consensus guidelines, on behalf of the Australasian Society of Thrombosis and Haemostasis. Med J Aust2004; 181:492–497.
Davis SM, Broderick J, Hennerici M, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology2006; 66:1175–1181.
Broderick JP, Diringer MN, Hill MD, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Determinants of intracerebral hemorrhage growth: an exploratory analysis. Stroke2007; 38:1072–1075.
Goldstein JN, Fazen LE, Snider R, et al. Contrast extravasation on CT angiography predicts hematoma expansion in intracerebral hemorrhage. Neurology2007; 68:889–894.
Kazui S, Naritomi H, Yamamoto H, Sawada T, Yamaguchi T. Enlargement of spontaneous intracerebral hemorrhage. Incidence and time course. Stroke1996; 27:1783–1787.
Kim J, Smith A, Hemphill JC, et al. Contrast extravasation on CT predicts mortality in primary intracerebral hemorrhage. AJNR Am J Neuroradiol2008; 29:520–525.
Wada R, Aviv RI, Fox AJ, et al. CT angiography “spot sign” predicts hematoma expansion in acute intracerebral hemorrhage. Stroke2007; 38:1257–1262.
Barras CD, Tress BM, Christensen S, et al; Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators. Density and shape as CT predictors of intracerebral hemorrhage growth. Stroke2009; 40:1325–1331.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. Systematic characterization of the computed tomography angiography spot sign in primary intracerebral hemorrhage identifies patients at highest risk for hematoma expansion: the spot sign score. Stroke2009; 40:2994–3000.
Fujii Y, Takeuchi S, Sasaki O, Minakawa T, Tanaka R. Multivariate analysis of predictors of hematoma enlargement in spontaneous intracerebral hemorrhage. Stroke1998; 29:1160–1166.
Ederies A, Demchuk A, Chia T, et al. Postcontrast CT extravasation is associated with hematoma expansion in CTA spot negative patients. Stroke2009; 40:1672–1676.
Delgado P, Alvarez-Sabín J, Abilleira S, et al. Plasma d-dimer predicts poor outcome after acute intracerebral hemorrhage. Neurology2006; 67:94–98.
Naidech AM, Jovanovic B, Liebling S, et al. Reduced platelet activity is associated with early clot growth and worse 3-month outcome after intracerebral hemorrhage. Stroke2009; 40:2398–2401.
Silva Y, Leira R, Tejada J, Lainez JM, Castillo J, Dávalos A; Stroke Project, Cerebrovascular Diseases Group of the Spanish Neurological Society. Molecular signatures of vascular injury are associated with early growth of intracerebral hemorrhage. Stroke2005; 36:86–91.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Moussouttas M, Malhotra R, Fernandez L, et al. Role of antiplatelet agents in hematoma expansion during the acute period of intracerebral hemorrhage. Neurocrit Care2010; 12:24–29.
Delgado Almandoz JE, Yoo AJ, Stone MJ, et al. The spot sign score in primary intracerebral hemorrhage identifies patients at highest risk of in-hospital mortality and poor outcome among survivors. Stroke2010; 41:54–60.
Broderick J, Connolly S, Feldmann E, et al; American Heart Association; American Stroke Association Stroke Council; High Blood Pressure Research Council; Quality of Care and Outcomes in Research Interdisciplinary Working Group. Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Stroke2007; 38:2001–2023.
Steiner T, Kaste M, Forsting M, et al. Recommendations for the management of intracranial haemorrhage - part I: spontaneous intracerebral haemorrhage. The European Stroke Initiative Writing Committee and the Writing Committee for the EUSI Executive Committee. Cerebrovasc Dis2006; 22:294–316.
Steiner T, Rosand J, Diringer M. Intracerebral hemorrhage associated with oral anticoagulant therapy: current practices and unresolved questions. Stroke2006; 37:256–262.
Goldstein JN, Fazen LE, Wendell L, et al. Risk of thromboembolism following acute intracerebral hemorrhage. Neurocrit Care2009; 10:28–34.
Christensen MC, Dawson J, Vincent C. Risk of thromboembolic complications after intracerebral hemorrhage according to ethnicity. Adv Ther2008; 25:831–841.
Lacut K, Bressollette L, Le Gal G, et al; VICTORIAh (Venous Intermittent Compression and Thrombosis Occurrence Related to Intracerebral Acute hemorrhage) Investigators. Prevention of venous thrombosis in patients with acute intracerebral hemorrhage. Neurology2005; 65:865–869.
Diringer MN, Skolnick BE, Mayer SA, et al. Thromboembolic events with recombinant activated factor VII in spontaneous intracerebral hemorrhage: results from the Factor Seven for Acute Hemorrhagic Stroke (FAST) trial. Stroke2010; 41:48–53.
Boeer A, Voth E, Henze T, Prange HW. Early heparin therapy in patients with spontaneous intracerebral haemorrhage. J Neurol Neurosurg Psychiatry1991; 54:466–467.
Dickmann U, Voth E, Schicha H, Henze T, Prange H, Emrich D. Heparin therapy, deep-vein thrombosis and pulmonary embolism after intracerebral hemorrhage. Klin Wochenschr1988; 66:1182–1183.
Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):381S–453S.
Vermeer SE, Algra A, Franke CL, Koudstaal PJ, Rinkel GJ. Long-term prognosis after recovery from primary intracerebral hemorrhage. Neurology2002; 59:205–209.
Bailey RD, Hart RG, Benavente O, Pearce LA. Recurrent brain hemorrhage is more frequent than ischemic stroke after intracranial hemorrhage. Neurology2001; 56:773–777.
Viswanathan A, Rakich SM, Engel C, et al. Antiplatelet use after intracerebral hemorrhage. Neurology2006; 66:206–209.
Greenberg SM, Eng JA, Ning M, Smith EE, Rosand J. Hemorrhage burden predicts recurrent intracerebral hemorrhage after lobar hemorrhage. Stroke2004; 35:1415–1420.
Lee SH, Ryu WS, Roh JK. Cerebral microbleeds are a risk factor for warfarin-related intracerebral hemorrhage. Neurology2009; 72:171–176.
Ueno H, Naka H, Ohshita T, et al. Association between cerebral microbleeds on T2*-weighted MR images and recurrent hemorrhagic stroke in patients treated with warfarin following ischemic stroke. AJNR Am J Neuroradiol2008; 29:1483–1486.
O’Donnell HC, Rosand J, Knudsen KA, et al. Apolipoprotein E genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med2000; 342:240–245.
Genes for Cerebral Hemorrhage on Anticoagulation (GOCHA) Collaborative Group. Exploiting common genetic variation to make anticoagulation safer. Stroke2009; 40(suppl 3):S64–S66.
Tzourio C, Arima H, Harrap S, et al. APOE genotype, ethnicity, and the risk of cerebral hemorrhage. Neurology2008; 70:1322–1328.
Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for predicting the risk of major bleeding in outpatients treated with warfarin. Am J Med1998; 105:91–99.
Kuijer PM, Hutten BA, Prins MH, Büller HR. Prediction of the risk of bleeding during anticoagulant treatment for venous thromboembolism. Arch Intern Med1999; 159:457–460.
Wess ML, Schauer DP, Johnston JA, et al. Application of a decision support tool for anticoagulation in patients with non-valvular atrial fibrillation. J Gen Intern Med2008; 23:411–417.
Singer DE, Chang Y, Fang MC, et al. Should patient characteristics influence target anticoagulation intensity for stroke prevention in nonvalvular atrial fibrillation? The ATRIA study. Circ Cardiovasc Qual Outcomes2009; 2:297–304.
Fang MC, Chang Y, Hylek EM, et al. Advanced age, anticoagulation intensity, and risk for intracranial hemorrhage among patients taking warfarin for atrial fibrillation. Ann Intern Med2004; 141:745–752.
Singer DE, Chang Y, Fang MC, et al. The net clinical benefit of warfarin anticoagulation in atrial fibrillation. Ann Intern Med2009; 151:297–305.
Eckman MH, Rosand J, Knudsen KA, Singer DE, Greenberg SM. Can patients be anticoagulated after intracerebral hemorrhage? A decision analysis. Stroke2003; 34:1710–1716.
Baruch L, Gage BF, Horrow J, et al. Can patients at elevated risk of stroke treated with anticoagulants be further risk stratified?Stroke2007; 38:2459–2463.
Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the National Registry of Atrial Fibrillation. JAMA2001; 285:2864–2870.
Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation1994; 89:635–641.
Freudenberger RS, Hellkamp AS, Halperin JL, et al; SCD-HeFT Investigators. Risk of thromboembolism in heart failure: an analysis from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation2007; 115:2637–2641.
Prandoni P, Lensing AW, Cogo A, et al. The long-term clinical course of acute deep venous thrombosis. Ann Intern Med1996; 125:1–7.
Claassen DO, Kazemi N, Zubkov AY, Wijdicks EF, Rabinstein AA. Restarting anticoagulation therapy after warfarin-associated intracerebral hemorrhage. Arch Neurol2008; 65:1313–1318.
De Vleeschouwer S, Van Calenbergh F, van Loon J, Nuttin B, Goffin J, Plets C. Risk analysis of thrombo-embolic and recurrent bleeding events in the management of intracranial haemorrhage due to oral anticoagulation. Acta Chir Belg2005; 105:268–274.
Butler AC, Tait RC. Restarting anticoagulation in prosthetic heart valve patients after intracranial haemorrhage: a 2-year follow-up. Br J Haematol1998; 103:1064–1066.
Bertram M, Bonsanto M, Hacke W, Schwab S. Managing the therapeutic dilemma: patients with spontaneous intracerebral hemorrhage and urgent need for anticoagulation. J Neurol2000; 247:209–214.
Taylor FC, Cohen H, Ebrahim S. Systematic review of long term anticoagulation or antiplatelet treatment in patients with nonrheumatic atrial fibrillation. BMJ2001; 322:321–326.
Flaherty ML, Tao H, Haverbusch M, et al. Warfarin use leads to larger intracerebral hematomas. Neurology2008; 71:1084–1089.
Sansing LH, Messe SR, Cucchiara BL, Cohen SN, Lyden PD, Kasner SE; CHANT Investigators. Prior antiplatelet use does not affect hemorrhage growth or outcome after ICH. Neurology2009; 72:1397–1402.
Snow V, Qaseem A, Barry P, et al; American College of Physicians; American Academy of Family Physicians Panel on Deep Venous Thrombosis/Pulmonary Embolism. Management of venous thromboembolism: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med2007; 146:204–210.
Albers GW, Amarenco P, Easton JD, Sacco RL, Teal PA; merican College of Chest Physicians. Antithrombotic and thrombolytic therapy for ischemic stroke: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):630S–639S.
Hankey GJ, Eikelboom JW. Antithrombotic drugs for patients with ischaemic stroke and transient ischaemic attack to prevent recurrent major vascular events. Lancet Neurol2010; 9:273–284.
Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009; 361:2342–2352.
Schulman S, Wåhlander K, Lundström T, Clason SB, Eriksson H; THRIVE III Investigators. Secondary prevention of venous thromboembolism with the oral direct thrombin inhibitor ximelagatran. N Engl J Med2003; 349:1713–1721.
Singer DE, Albers GW, Dalen JE, et al; American College of Chest Physicians. Antithrombotic therapy in atrial fibrillation: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest2008; 133(suppl 6):546S–592S.
Given the high risk of hematoma expansion in the early phase of acute ICH, most experts recommend reversing anticoagulation immediately.
Many clinicians start subcutaneous heparinoids in low doses 24 to 72 hours after ICH to prevent deep vein thrombosis, and after the first few days or a week, consider either increasing the dose to a full anticoagulation dose or making a transition to oral anticoagulants.
Many patients with lobar hemorrhage or cerebral amyloid angiopathy may remain at higher risk of anticoagulant-related ICH recurrence than thromboembolic events and would therefore be best managed without anticoagulants.
Those with deep hemispheric ICH, hypertension that can be well controlled, and a high risk of disabling thromboembolism may receive net benefit from restarting anticoagulation.
Hyponatremia, defined as a serum sodium concentration below 135 mmol/L, is one of the most frequently encountered electrolyte disorders. In 1981, Flear et al1 reported that 15% of their hospitalized patients had plasma sodium concentrations lower than 134 mmol/L, the cutoff they were using at that time.
Hyponatremia is sometimes merely a laboratory artifact or a result of improper blood collection. If real, it can be due to excessive water intake or, most often, the inability of the kidney to excrete water coupled with continued water intake. Patients with significant underlying cardiac, hepatic, or renal dysfunction are at greatest risk of developing hyponatremia, secondary to the nonosmotic release of antidiuretic hormone (ADH). Others at risk include postoperative patients (especially menstruating women), older patients on thiazide diuretics, patients with malignant or psychiatric illness, and endurance athletes.
In this article, we review the treatment of acute and chronic hyponatremia, emphasizing the importance of basing the therapy on the severity of symptoms and taking care not to raise the serum sodium level too rapidly, which can cause neurologic dysfunction.
Guidelines for managing hyponatremia2 are based mostly on retrospective studies and expert opinion, since few prospective studies have been done. Despite the paucity of evidence-based recommendations, we will attempt to incorporate findings from important human and animal studies and consensus guidelines from expert panels. We will focus initially on the critical diagnostic considerations necessary to initiate treatment.
SYMPTOMATIC VS ASYMPTOMATIC
Subsequent sections will address therapeutic approaches in two clinical settings:
Symptomatic hyponatremia, ie, with severe signs or symptoms of cerebral edema—a medical emergency; and
Asymptomatic hyponatremia, ie, without serious signs or symptoms of cerebral edema.
KEY DIAGNOSTIC STEPS WHEN STARTING TREATMENT
The treatment of hyponatremia begins by confirming a truly hypo-osmolar state, assessing its clinical significance, and determining its cause (Table 1).
The clinical and laboratory evaluations form the foundation of a proper approach to any patient with hyponatremia. The rationale behind making several important diagnostic distinctions will be discussed here briefly and then expanded on in the remaining text. The reader is referred to another review on the diagnostic evaluation of hyponatremia.3
Confirm that the patient truly has hypo-osmolar hyponatremia
The serum osmolality should be measured to confirm that it is low (ie, < 275 mOsm/kg). In addition, the arterial serum sodium concentration can be measured using a blood gas device if pseudohyponatremia (see below) is suspected. This method uses direct potentiometry and bypasses the dilutional step in the processing of venous samples.4
Rationale. The clinical consequences of hyponatremia are due to water moving from hypo-osmolar extracellular fluid into the relatively hyperosmolar interior of the cell. This water movement can cause progressive cerebral edema, resulting in a spectrum of signs and symptoms from headache and ataxia to seizures and coma. But significant fluid shifts and cerebral edema occur only if the extracellular fluid is hypo-osmolar relative to the intracellular fluid.
In fact, hyponatremia can occur in several situations in which the extracellular fluid is not hypo-osmolar. An increase in effective plasma osmoles (substances in the extracellular fluid that do not readily move across the plasma membrane) can cause water to move out of cells, resulting in translocational hyponatremia. This may be seen in hyperglycemia or when mannitol or contrast dye has been given. In these situations, the plasma is either isotonic or even hypertonic to the intracellular fluid, resulting in no movement of water into the cells and therefore no clinical consequences relating to the hyponatremia. Importantly, no therapy is required for the hyponatremia.
Other situations in which hyponatremia is present but not associated with true hypotonicity include states of excess protein or lipid in the blood (pseudohyponatremia). Also, if an infusion of hypotonic fluid is running, clinicians must be sure that blood samples are not drawn proximally in the same vein.
Are there significant signs or symptoms of cerebral edema?
Hyponatremia can cause brain swelling within the confined space of the skull as water shifts from the extracellular fluid into the cells. Depending on underlying risk factors (Table 2)5 and the severity and duration of hyponatremia (see below), this may result in signs or symptoms of cerebral edema, including visual changes, focal neurologic changes, encephalopathy, respiratory depression, and seizures. Ultimately, brain herniation can occur.
Patients need to be assessed quickly because those with serious neurologic signs or symptoms thought to be related to hyponatremia require urgent treatment with hypertonic saline to increase the serum sodium concentration, regardless of the underlying volume status, the cause of hyponatremia, or the time of onset.
Determine the duration of hyponatremia
One should try to ascertain when the hyponatremia started, as its duration is important in determining the proper pace of correction.
Figure 1.The brain begins to adapt to hyponatremia within minutes, and the cerebral adaptation is maximal within 2 to 3 days (Figure 1).6
At the onset of hyponatremia, water moves from the extracellular fluid into cells, pulled in by osmosis. The brain can decrease the net amount of water entering into the neurons (and thus regulate its volume) by increasing the flow of water from the interstitium into the cerebrospinal fluid via increased interstitial hydraulic pressure.7
Over the next several days, inorganic solutes (eg, potassium and sodium salts) and various organic solutes are transported out of the cells. In patients in whom this process has had time to occur, treatment of hyponatremia with hypertonic fluids raises the plasma osmolality faster than the cells can recapture the previously transported osmoles. In this situation, overly rapid correction can cause excessive loss of intracellular water, resulting in cell shrinkage and osmotic demyelination syndrome. Osmotic demyelination usually presents during treatment of hyponatremia after an initial improvement in mental status, with worsening neurologic function and various neurologic signs, including paresis and ultimately even death.6
In patients with acute-onset hyponatremia (ie, with onset within the past 48 hours), in whom the above cerebral adaptations have not had time to occur completely, rapid correction is unlikely to result in osmotic demyelination.
In view of the serious risk of osmotic demyelination, if the timing of development of hyponatremia cannot be determined, one should assume it is chronic (> 48 hours) and avoid rapid overcorrection (see discussion below on the rate of correction).
On the other hand, patients who have severe neurologic signs or symptoms initially need their serum sodium increased urgently to safer levels, regardless of the timing of onset (see below for suggested approach). Subsequent treatment of hyponatremia—after the serum sodium level has been raised enough to reverse neurologic symptoms—will be influenced by the duration of the hyponatremia, with careful avoidance of overly rapid correction, especially in patients with chronic hyponatremia.
Assess the patient’s volume status to determine the proper initial treatment
In patients with hypo-osmolar hyponatremia who do not need urgent therapy with hypertonic saline, the initial treatment is based on clinical and laboratory assessment of extracellular fluid volume status, including spot urine sodium measurement (Table 3).3 This will be discussed further below.
Check urine osmolality to assess for hyponatremic states in which urinary dilution is intact
Measuring urine osmolality is useful in ascertaining whether hyponatremic patients are making appropriately dilute urine (< 100 mOsm/kg). If they are, the cause of the hyponatremia may be excessive water intake, a reset osmostat, or low solute intake. In addition, patients with hypovolemic hyponatremia may have appropriately dilute urine soon after treatment with isotonic intravenous fluids.
The serum sodium concentration often returns to normal if the underlying cause is eliminated (eg, if excessive fluid intake is stopped). If there are no serious signs or symptoms, this can usually be accomplished without additional therapy with intravenous fluids or medications, thereby avoiding the risk of overcorrection.
Search for causes of rapidly correctable hyponatremia
If hyponatremia is due to one of several important underlying causes, it may reverse rapidly once the underlying cause has been eliminated (Table 4). Examples: restricting water intake in patients with psychogenic polydipsia, discontinuing thiazide diuretics, replenishing depleted fluid volume, stopping desmopressin (DDAVP), and giving glucocorticoid replacement to those who are glucocorticoid-deficient.
TREATING HYPONATREMIC PATIENTS WITH SERIOUS SIGNS OR SYMPTOMS
Patients with hypo-osmolar hyponatremia and serious signs or symptoms of cerebral edema (lethargy, respiratory depression, seizures) need rapid initial correction of the serum sodium level, as this is a true medical emergency.
Certain patients are at greater risk of developing cerebral edema from hyponatremia (Table 2).
On the other hand, patients with chronic hyponatremia are very unlikely to present with signs or symptoms of cerebral edema. In fact, in a patient with chronic hyponatremia, care must be taken to avoid overcorrection beyond that needed to reverse severe signs and symptoms. In the rare case in which a patient with chronic hyponatremia presents with signs or symptoms of cerebral edema, the hypertonic saline infusion must be stopped as soon as the signs or symptoms have resolved. Further rapid changes in serum sodium must be avoided.
During correction of hyponatremia, some patients are at particularly high risk of osmotic demyelination syndrome secondary to underlying abnormalities in cerebral osmotic regulation. These include patients with alcoholism, malnutrition, hypokalemia, and burns, and elderly women on thiazide diuretics.8 These patients should be monitored vigilantly for overly rapid correction during treatment.
INITIAL TREATMENT: REVERSE CEREBRAL EDEMA WITH 3% SALINE
The goal of the initial, rapid phase of correction is to reverse cerebral edema.
Patients with serious signs or symptoms should receive hypertonic (3%) saline at a rate of about 1 mL/kg/hour for the first several hours.8 Those with concomitant hypervolemia (as in congestive heart failure) or underlying cardiovascular disease should also receive a loop diuretic to aid in free-water excretion and to prevent volume overload from the saline infusion. This regimen usually raises the serum sodium concentration enough (usually by about 1 mmol/L/hour) to decrease cerebral edema and improve symptoms.
In patients having active seizures or showing signs of brain herniation, 3% saline can be given initially at a higher rate of about 2 to 3 mL/kg/hour over the first few hours. An alternative approach is an initial 50-mL bolus of 3% saline and an additional 200 mL given over the subsequent 4 to 6 hours.9
No study has compared the efficacy and safety of these approaches, and clinicians should always monitor extracellular fluid volume status, neurologic status, and serum sodium levels closely in any patient treated with hypertonic saline.
After severe signs and symptoms have resolved, 3% saline is promptly discontinued and appropriate therapy is initiated based on the patient’s volume status and underlying cause of hyponatremia (see discussion below).
NEXT, FIND THE APPROPRIATE RATE OF CORRECTION
After the initial serious signs or symptoms have been addressed with hypertonic saline, management should focus on limiting the rate of correction in patients with chronic hyponatremia or hyponatremia of unknown duration.
Animal studies and retrospective human studies have suggested certain guidelines on the appropriate pace and magnitude of correction during treatment of hyponatremia to avoid osmotic demyelination syndrome.2
Clinicians must not attempt to correct the serum sodium to “normal” values. Although patients with acute hyponatremia may tolerate complete correction, there is little evidence that raising the serum sodium concentration acutely by more than 5 to 8 mmol/L is advantageous. Therefore, correction should be judicious in all patients.
Appropriate rates of correction
A recent expert consensus panel suggested that the serum sodium level be raised by no more than 10 to 12 mmol/L during the first 24 hours of treatment, and by less than 18 mmol/L over 48 hours.2
Patients with chronic hyponatremia and signs or symptoms of cerebral edema should have their sodium level raised at an even slower rate—some recommend less than 10 mmol/L in the first 24 hours.10 Aggressive initial correction at the rate of 1.5 to 2 mmol/hour for the first 3 to 4 hours with 3% saline is indicated until serious symptoms (seizure, obtundation) resolve, but correction beyond 10 to 12 mmol/L in the first 24 hours should be avoided. Hypertonic saline therapy should usually be discontinued well before the serum sodium level has risen this much, to avoid a continuing rise in the sodium level after the infusion has stopped.
While hypertonic saline is being infused, serum sodium levels should be checked every 1 to 2 hours. In a study in 56 patients with severe hyponatremia (serum sodium ≤ 105 mmol/L),11 no neurologic complications were observed in patients with chronic hyponatremia whose serum sodium was corrected by less than 12 mmol/L in 24 hours or by less than 18 mmol/L in 48 hours or in whom the average rate of correction to a serum sodium of 120 mmol/L was less than or equal to 0.55 mmol/L per hour.
If the serum sodium concentration has been overcorrected
Desmopressin is effective in preventing and reversing inadvertent overcorrection of hyponatremia. 12 In one study, desmopressin lowered the sodium concentration by 2 to 9 mmol/L in 14 of 20 patients. None of the patients developed any serious adverse consequences.
In addition, intravenous water (dextrose 5%) can be given alone or in combination with desmopressin to prevent or reverse an excessive increase in serum sodium.13 Such therapy may be considered in patients who continue to excrete hypotonic urine and have already reached a serum sodium concentration that meets or exceeds the recommended rate or magnitude of change.
Formulas for estimating the rate of correction
Various formulas have been devised for estimating the change in serum sodium concentration during treatment of hyponatremia.14
The Adrogué-Madias formula, one of the most commonly used, gives an estimate of how much the serum sodium concentration will rise when 1 L of various intravenous fluids is given (Table 5).15 This formula also accounts for the increase in serum sodium that takes place during concomitant correction of hypokalemia with potassium. Recently, however, a retrospective study16 found that this formula underestimated the change in serum sodium in 23 (74.2%) of 31 patients with hyponatremia treated with hypertonic saline.
An alternative is the Barsoum-Levine equation, which takes into account ongoing urinary losses. Although it is more cumbersome to calculate, it may be more precise.17
Alternatively, in patients without hypovolemia, the clinician can calculate the amount of urinary excretion of free water required to achieve a specific target serum sodium and then measure hourly urinary water excretion during aquaresis induced by furosemide (Lasix).8 Although more physiologic, this method can be clinically cumbersome, requiring timely handling of urine specimens, accurate recording of urine output, and rapid reporting of laboratory results.
Ultimately, these methods serve only as estimates of the change in serum sodium and do not replace careful monitoring of electrolytes (every 1 to 2 hours during acute therapy) and fastidious assessment for clinical signs or symptoms of osmotic demyelination syndrome.
PATIENTS WITH HYPONATREMIA AND NO SERIOUS SIGNS OR SYMPTOMS
General approach
Hyponatremic patients without serious signs or symptoms of cerebral edema do not require urgent therapy to raise the serum sodium.
Patients with chronic asymptomatic hyponatremia are commonly encountered in clinical practice. As a result of cerebral adaptation, they can appear to have no symptoms despite serum sodium levels as low as 115 to 120 mmol/L. However, even if they have no serious signs or symptoms of cerebral edema, some patients may complain of fatigue, lethargy, nausea, gait abnormalities, and muscle cramps and have evidence of milder forms of neurocognitive impairment.18
In a recent case-control study,18 elderly patients with chronic hyponatremia (mean serum sodium concentration 126 ± 5 mmol/L) were more likely to present to the hospital with falls compared with age-matched controls. Further analysis suggested these patients had marked impairments in gait and attention, which improved in some as the serum sodium increased.
Another recent study19 reported that mild hyponatremia (mean serum sodium concentration 132 mmol/L) was independently associated with the risk of fracture, even after adjustment for known osteoporotic risk factors.
Even when there is no need for acute therapy to raise the serum sodium level, the clinician should scrutinize the medical regimen and available clinical data to rule out reversible causes of water excess. These may include ongoing administration of hypotonic fluids (eg, parenteral nutrition or dextrose 5% to “keep the vein open”) or of medications that cause inappropriate release of ADH (eg, selective serotonin reuptake inhibitors) or that impair water excretion (eg, nonsteroidal anti-inflammatory drugs). The clinician should also search for an underlying diagnosis that predisposes to water retention, such as hypothyroidism, adrenal insufficiency, congestive heart failure, or hepatic or renal failure. If hyponatremia is due to endocrine disease, correction of hypothyroidism or adrenal insufficiency should result in water excretion and improvement in the serum sodium.
If the cause of the hyponatremia is not immediately apparent, treatment can be started on the basis of assessment of the patient’s extracellular fluid volume status using clinical examination and supplementary laboratory data such as the serum uric acid concentration and urinary sodium concentration.3Table 3 outlines general treatment options for hypoosmolar hyponatremia according to extracellular fluid volume status.
Of note, physical examination alone has poor sensitivity and specificity in assessing extracellular fluid volume status in patients with hyponatremia.20,21 This highlights the importance of spot measurements of urine sodium and serum uric acid and, when appropriate, isotonic intravenous saline challenge to detect occult hypovolemia.
In general, patients with euvolemia are treated with fluid restriction, and patients with hypovolemia are given isotonic saline. Patients with hypervolemia can be difficult to treat, but in general they are prescribed both sodium and fluid restriction. Loop diuretics can be given to promote excretion of water and sodium. Thiazide diuretics are avoided, as they impair urinary dilution and worsen hyponatremia. Attempts should be made to optimize the treatment of the underlying hypervolemic disorder (congestive heart failure, cirrhosis, advanced renal failure). Vasopressin receptor antagonists can also be used in selected cases of hypervolemic or euvolemic hyponatremia (see discussion below).
How to prescribe fluid restriction rationally
Ideally, patients should not ingest any more fluid than they can excrete in urine and insensible losses—otherwise, the serum sodium can continue to decrease.
Water excretion can be estimated from solute intake and urine osmolarity. In theory, a 70-kg person with a typical daily solute intake of about 10 mOsm/kg and intact urinary dilution to a urine osmolarity of 50 mOsm/L can excrete up to 14 L of urine (700 mOsm/50 mOsm/L) per day. However, a patient with the syndrome of inappropriate ADH secretion (SIADH) and a fixed urine osmolality of 700 mOsm/kg would excrete a similar solute load in only 1 L of urine. Thus, any fluid intake in excess of this volume could worsen hyponatremia.
To excrete free water, urinary sodium plus urinary potassium must be less than the serum sodium concentration. In this regard, the necessary degree of fluid restriction can also be estimated made on the basis of the patient’s urinary electrolytes.22
Increased solute intake to augment water excretion
In patients without hypervolemia, solute intake can be increased to augment water excretion. 22 This can be achieved with salt tablets or oral urea. Although urea can be effective, it is not commonly used because it is not available the United States and it has poor gastrointestinal tolerability. In patients whose nutritional intake is limited and who continue to ingest fluids (such as, for example, an elderly patient subsisting on tea and toast) every effort should be made to increase solute intake with high-protein foods or supplements.
DRUGS TO INHIBIT VASOPRESSIN
Unfortunately, patients often do not adhere to these strategies, as fluid restriction and unpalatable salt tablets or urea can become too burdensome. In such instances, pharmacologic inhibition of vasopressin-mediated water reabsorption can be considered using the following agents.
Demeclocycline (Declomycin) and lithium inhibit the kidney’s response to vasopressin. Because lithium may be nephrotoxic and has unwanted effects on the central nervous system, demeclocycline has become the preferred agent. Given in doses of 300 to 600 mg twice daily, demeclocycline promotes free water excretion, but often takes 1 to 2 weeks of therapy to begin working.
Renal failure due to demeclocycline has been reported in patients with concomitant liver disease.23 Demeclocycline can also cause photosensitivity and is contraindicated in children and pregnant women due to abnormalities in bone and enamel formation. In addition, it can be expensive and may not be covered fully by some prescription plans.
Vasopressin receptor antagonists (‘vaptans’)
ADH, also called vasopressin, interacts with various receptor subtypes, including V1a (causing vasoconstriction, platelet aggregation, inotropic stimulation, myocardial protein synthesis), V1b (causing secretion of adrenocorticotropic hormone), and V2 (causing water reabsorption and release of von Willebrand factor and factor VIII).
Drugs that block V2 receptors in the renal tubule increase water excretion, making them attractive as therapy for some hyponatremic states (Table 6).24,25 These drugs exert their aquaretic effect by causing a decrease in transcription and insertion of aquaporin-2 channels (“water pores”) into the apical collecting duct membrane. As a result, the water permeability of the collecting duct is decreased even in the presence of circulating ADH.
Conivaptan (Vaprisol) is a combined V1a-V2 antagonist that has been approved for the treatment of euvolemic and hypervolemic hyponatremia. Conivaptan inhibits the cytochrome P450 3A4 system and thus may interact with other drugs; therefore, its use has been limited to no more than 4 days of intravenous administration in the hospital setting. The recommended dosage is an initial 20-mg infusion over 30 minutes, followed by continuous infusions of 20 to 40 mg/day. Dosing adjustments in renal and hepatic impairment have not been well defined.
Tolvaptan (Samsca) is an oral selective V2 antagonist that has been studied in patients with euvolemic and hypervolemic hyponatremia. 26 Studies have included patients with congestive heart failure, cirrhosis, and SIADH. Although tolvaptan has not been shown to reduce rates of rehospitalization or death in congestive heart failure, it improves serum sodium, overall fluid balance, and congestive symptoms.27 Tolvaptan has recently been approved for the treatment of euvolemic and hypervolemic hyponatremia.
A recent study has confirmed the longterm efficacy of tolvaptan in 111 patients over a mean duration of treatment greater than 700 days.28 While the clinical benefits of chronic tolvaptan therapy have yet to be clearly demonstrated, this study shows that tolvaptan therapy can result in a sustained improvement in serum sodium concentration without an unacceptable increase in adverse events.29
Lixivaptan (VPA-985), another oral selective V2 receptor antagonist, is being studied in patients with euvolemic and hypervolemic hyponatremia.
Current role of vasopressin antagonists
Current studies of vasopressin antagonists in the treatment of hyponatremia are promising, though definite recommendations are needed to ensure slow, careful correction of hyponatremia. Most studies suggest that these agents provide slow, reliable increases in serum sodium. In one large study of patients with congestive heart failure, serum sodium rose by more than 12 mmol/L in 24 hours in fewer than 2% of patients.26
Notably, no cases of osmotic demyelination syndrome have been reported in these studies. However, it should be noted that therapy was started in the hospital with close monitoring of serum sodium levels and discontinuation of fluid restriction; the incidence of overly rapid correction of sodium may be higher outside of carefully done clinical studies. Clinicians should adopt monitoring strategies similar to those used in these rigorous studies.
At present, there is little experience with vasopressin antagonists in hyponatremic patients with serious signs or symptoms of cerebral edema, and most clinicians still view 3% saline as the gold standard for these patients.
Vasopressin antagonists should not be used in patients with hypovolemic hyponatremia, due to concerns about V1a blockade causing hypotension and about V2 blockade producing water excretion and a worsening of the volume-depleted state.
Recent clinical trials have reported that patients often experience increased thirst while taking these agents. This highlights the need to monitor serum sodium during treatment.
These agents are expensive. Tolvaptan costs about $250 per tablet; conivaptan, which is administered intravenously, may cost a little more per treatment course.
THERAPY IN SPECIFIC DISEASE STATES
Patients with hyponatremia and cirrhosis
The focus of treatment remains water and salt restriction and judicious use of loop diuretics and aldosterone antagonists such as spironolactone (Aldactone).
Tolvaptan has been effective at raising the serum sodium level in patients with cirrhosis, 26 while conivaptan should be avoided at present because of vasodilation from V1a receptor antagonism and its potential effects on systemic hemodynamics and risk of variceal bleeding.30
As the severity of cirrhosis increases, the only effective treatment of hyponatremia is liver transplantation.
Patients with SIADH
In most cases, water restriction is the mainstay of therapy. Adequate nutritional intake should also be stressed so that enough solute is available for ongoing water excretion. Although fluid restriction is usually effective, many patients cannot adhere to the level of restriction required.
In cases in which fluid restriction is not effective on its own, demeclocyline can be used to antagonize ADH action and increase water excretion. Sodium tablets and loop diuretics can also be used, taking care to avoid hypovolemia from diuretic-induced sodium losses. The use of tolvaptan in patients with SIADH has resulted in short-term increases in serum sodium.26 A recent study has suggested that this effect can be sustained with longer-term treatment,28 but further studies are needed to show a complementary clinical benefit (eg, improved neurocognition) to guide the use of these costly agents in clinical practice.
Patients with diuretic-induced hyponatremia
Thiazide diuretics should be discontinued and hypovolemia and hypokalemia should be corrected with isotonic saline and potassium supplementation. As the hypokalemia is corrected and the diuretic effect and hypovolemic stimulus to ADH dissipates, water excretion can increase rapidly, resulting in a brisk change in serum sodium.
Serum sodium levels should be closely monitored during therapy to avoid overcorrection. For this reason, use of hypertonic saline should generally be avoided. Hypotonic fluid— eg, half-normal (0.45%) or quarter-normal (0.22%) saline or even desmopressin—may become necessary in the later stages of therapy to avoid overly rapid correction.
Patients with exercise-associated hyponatremia
Patients at highest risk of exercise-associated hyponatremia include those who drink too much fluid during a long-distance race, who have low body weight, who are female, who exercise longer than 4 hours, and who use nonsteroidal anti-inflammatory drugs.31 The cause of hyponatremia is likely multifactorial, with excessive water intake coupled with sodium losses and impaired renal excretion of water due to ADH action and impaired renal dilution. To prevent exercise-associated hyponatremia, fluid intake should be limited to 400 to 800 mL/hour, with the higher end recommended for larger athletes and hotter climates.
Consensus recommendations suggest that most patients with mild hyponatremia (serum sodium 130 to 135 mmol/L) should be treated with fluid restriction and clinical observation, as spontaneous water diuresis leads to improvement in the serum sodium level. Importantly, the reflex to provide isotonic saline infusions should be avoided unless clear signs of volume depletion are present. Intravenous saline has the potential to worsen hyponatremia in the presence of ADH. In addition, some athletes will have retained water in the gastrointestinal tract that may be mobilized after the race, resulting in worsening of hyponatremia.32
In athletes with severe hyponatremia (serum sodium < 120 mmol/L) or symptomatic exercise-associated hyponatremia (lethargy, respiratory depression, seizures), hypertonic saline is the treatment of choice. One protocol suggests giving 100 mL of 3% saline over 10 minutes in the field, followed by prompt transportation to hospital.33
SUMMARY POINTS
Hyponatremia is a common electrolyte disorder that in its most severe form requires urgent therapy with hypertonic saline to correct cerebral edema.
In patients without serious signs or symptoms of cerebral edema, recent observations suggest there may be clinically important symptomatology relating to mild neurocognitive dysfunction and an association with risk of bone fracture.
Multiple treatment strategies are available according to the underlying extracellular fluid volume status and cause of hyponatremia. These include fluid and sodium restriction and augmentation of urinary water excretion with various nutritional and pharmacologic strategies. The most novel therapy includes antagonism of the vasopressin V2 receptor with a class of aquaretic agents known as vaptans.
There can be serious neurologic injury associated with overly rapid correction of chronic hyponatremia or undercorrection of acute symptomatic hyponatremia.
Clinicians must be familiar with the details of each of the treatments and have an appreciation of the importance of careful monitoring during treatment.
References
Flear CTG, Gill GV, Burn J. Hyponatremia: mechanisms and management. Lancet1981; 2:26–31.
Freda BJ, Davidson MB, Hall PM. Evaluation of hyponatremia: a little physiology goes a long way. Cleve Clin J Med2004; 71:639–650.
Weisberg LS. Pseudohyponatremia: a reappraisal. Am J Med1989; 86:315–318.
Moritz L, Ayus JC. The pathophysiology and treatment of hyponatraemic encephalopathy: an update. Nephrol Dial Transplant2003; 18:2486–2491.
Widdess-Walsh P, Sabharwal V, Demirjian S, DeGeorgia M. Neurologic effects of hyponatremia and its treatment. Cleve Clin J Med2007; 74:377–383.
Melton JE, Patlak CS, Pettigrew KD, Cserr HF. Volume regulatory loss of Na, Cl, and K from rat brain during acute hyponatremia. Am J Physiol1987; 252:F661–F669.
Lauriat SM, Berl T. The hyponatremic patient: practical focus on therapy. J Am Soc Nephrol1997; 8:1599–1607.
Kokko JP. Symptomatic hyponatremia with hypoxia is a medical emergency. Kidney Int2006; 69:1291–1293.
Ellis SJ. Severe hyponatraemia: complications and treatment. QJM1995; 88:905–909.
Sterns RH, Cappuccio JD, Silver SM, Cohen EP. Neurologic sequelae after treatment of severe hyponatremia: a multicenter perspective. J Am Soc Nephrol1994; 4:1522–1530.
Perianayagam A, Sterns RH, Silver SM, et al. DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia. Clin J Am Soc Nephrol2008; 3:331–336.
Sterns RH, Hix JK. Overcorrection of hyponatremia is a medical emergency. Kidney Int2009; 76:587–589.
Nguyen MK, Kurtz I. Analysis of current formulas used for treatment of the dysnatremias. Clin Exp Nephrol2004; 8:12–16.
Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med2000; 342:1581–1589.
Mohmand HK, Issa D, Ahmad Z, Cappuccio JD, Kouides RW, Sterns RH. Hypertonic saline for hyponatremia: risk of inadvertent overcorrection. Clin J Am Soc Nephrol2007; 2:1110–1117.
Ellison DH, Berl T. Clinical practice. The syndrome of inappropriate antidiuresis. N Engl J Med2007; 356:2064–2072.
Renneboog B, Musch W, Vandemergel X, Manto MU, Decaux G. Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits. Am J Med2006; 119:71.e1–e8.
Kinsella S, Moran S, Sullivan MO, Molloy MG, Eustace JA. Hyponatremia independent of osteoporosis is associated with fracture occurrence. Clin J Am Soc Nephrol2010; 5:275–280.
Chung HM, Kluge R, Schrier RW, Anderson RJ. Clinical assessment of extracellular fluid volume in hyponatremia. Am J Med1987; 83:905–988.
Hoorn EJ, Halperin ML, Zietse R. Diagnostic approach to a patient with hyponatraemia: traditional versus physiology-based options. QJM2005; 98:529–540.
Berl T. Impact of solute intake on urine flow and water excretion. J Am Soc Nephrol2008; 19:1076–1078.
Carrilho F, Bosch J, Arroyo V, Mas A, Viver J, Rodes J. Renal failure associated with demeclocycline in cirrhosis. Ann Intern Med1977; 87:195–197.
Lehrich RW, Greenberg A. When is it appropriate to use vasopressin receptor antagonists?J Am Soc Nephrol2008; 19:1054–1058.
Decaux G, Soupart A, Vassart G. Non-peptide arginine-vasopressin antagonists: the vaptans. Lancet2008; 371:1624–1632.
Schrier RW, Gross P, Gheorghiade M, et al; SALT Investigators. Tolvaptan, a selective oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl J Med2006; 355:2099–2112.
Konstam MA, Gheorghiade M, Burnett JC, et al; Efficacy of Vasopressin Antagonism in Heart Failure Outcome Study With Tolvaptan (EVEREST) Investigators. Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA2007; 297:1319–1331.
Berl T, Quittnat-Pelletier F, Verbalis JG, et al; SALTWATER Investigators. Oral tolvaptan is safe and effective in chronic hyponatremia. J Am Soc Nephrol2010; 21:705–712.
Greenberg A, Lehrich RW. Treatment of chronic hyponatremia: now we know how, but do we know when or if?J Am Soc Nephrol2010; 21:552–555.
Greenberg A, Verbalis JG. Vasopressin receptor antagonists. Kidney Int2006; 69:2124–2130.
Rosner MH, Kirven J. Exercise-associated hyponatremia. Clin J Am Soc Nephrol2007; 2:151–161.
Halperin ML, Kamel KS, Sterns R. Hyponatremia in marathon runners. N Engl J Med2005; 353:427–428.
Hew-Butler T, Almond C, Ayus JC, et al; Exercise-Associated Hyponatremia (EAH) Consensus Panel. Consensus statement of the 1st International Exercise-Associated Hyponatremia Consensus Development Conference, Cape Town, South Africa 2005. Clin J Sport Med2005; 15:208–213.
Hyponatremia, defined as a serum sodium concentration below 135 mmol/L, is one of the most frequently encountered electrolyte disorders. In 1981, Flear et al1 reported that 15% of their hospitalized patients had plasma sodium concentrations lower than 134 mmol/L, the cutoff they were using at that time.
Hyponatremia is sometimes merely a laboratory artifact or a result of improper blood collection. If real, it can be due to excessive water intake or, most often, the inability of the kidney to excrete water coupled with continued water intake. Patients with significant underlying cardiac, hepatic, or renal dysfunction are at greatest risk of developing hyponatremia, secondary to the nonosmotic release of antidiuretic hormone (ADH). Others at risk include postoperative patients (especially menstruating women), older patients on thiazide diuretics, patients with malignant or psychiatric illness, and endurance athletes.
In this article, we review the treatment of acute and chronic hyponatremia, emphasizing the importance of basing the therapy on the severity of symptoms and taking care not to raise the serum sodium level too rapidly, which can cause neurologic dysfunction.
Guidelines for managing hyponatremia2 are based mostly on retrospective studies and expert opinion, since few prospective studies have been done. Despite the paucity of evidence-based recommendations, we will attempt to incorporate findings from important human and animal studies and consensus guidelines from expert panels. We will focus initially on the critical diagnostic considerations necessary to initiate treatment.
SYMPTOMATIC VS ASYMPTOMATIC
Subsequent sections will address therapeutic approaches in two clinical settings:
Symptomatic hyponatremia, ie, with severe signs or symptoms of cerebral edema—a medical emergency; and
Asymptomatic hyponatremia, ie, without serious signs or symptoms of cerebral edema.
KEY DIAGNOSTIC STEPS WHEN STARTING TREATMENT
The treatment of hyponatremia begins by confirming a truly hypo-osmolar state, assessing its clinical significance, and determining its cause (Table 1).
The clinical and laboratory evaluations form the foundation of a proper approach to any patient with hyponatremia. The rationale behind making several important diagnostic distinctions will be discussed here briefly and then expanded on in the remaining text. The reader is referred to another review on the diagnostic evaluation of hyponatremia.3
Confirm that the patient truly has hypo-osmolar hyponatremia
The serum osmolality should be measured to confirm that it is low (ie, < 275 mOsm/kg). In addition, the arterial serum sodium concentration can be measured using a blood gas device if pseudohyponatremia (see below) is suspected. This method uses direct potentiometry and bypasses the dilutional step in the processing of venous samples.4
Rationale. The clinical consequences of hyponatremia are due to water moving from hypo-osmolar extracellular fluid into the relatively hyperosmolar interior of the cell. This water movement can cause progressive cerebral edema, resulting in a spectrum of signs and symptoms from headache and ataxia to seizures and coma. But significant fluid shifts and cerebral edema occur only if the extracellular fluid is hypo-osmolar relative to the intracellular fluid.
In fact, hyponatremia can occur in several situations in which the extracellular fluid is not hypo-osmolar. An increase in effective plasma osmoles (substances in the extracellular fluid that do not readily move across the plasma membrane) can cause water to move out of cells, resulting in translocational hyponatremia. This may be seen in hyperglycemia or when mannitol or contrast dye has been given. In these situations, the plasma is either isotonic or even hypertonic to the intracellular fluid, resulting in no movement of water into the cells and therefore no clinical consequences relating to the hyponatremia. Importantly, no therapy is required for the hyponatremia.
Other situations in which hyponatremia is present but not associated with true hypotonicity include states of excess protein or lipid in the blood (pseudohyponatremia). Also, if an infusion of hypotonic fluid is running, clinicians must be sure that blood samples are not drawn proximally in the same vein.
Are there significant signs or symptoms of cerebral edema?
Hyponatremia can cause brain swelling within the confined space of the skull as water shifts from the extracellular fluid into the cells. Depending on underlying risk factors (Table 2)5 and the severity and duration of hyponatremia (see below), this may result in signs or symptoms of cerebral edema, including visual changes, focal neurologic changes, encephalopathy, respiratory depression, and seizures. Ultimately, brain herniation can occur.
Patients need to be assessed quickly because those with serious neurologic signs or symptoms thought to be related to hyponatremia require urgent treatment with hypertonic saline to increase the serum sodium concentration, regardless of the underlying volume status, the cause of hyponatremia, or the time of onset.
Determine the duration of hyponatremia
One should try to ascertain when the hyponatremia started, as its duration is important in determining the proper pace of correction.
Figure 1.The brain begins to adapt to hyponatremia within minutes, and the cerebral adaptation is maximal within 2 to 3 days (Figure 1).6
At the onset of hyponatremia, water moves from the extracellular fluid into cells, pulled in by osmosis. The brain can decrease the net amount of water entering into the neurons (and thus regulate its volume) by increasing the flow of water from the interstitium into the cerebrospinal fluid via increased interstitial hydraulic pressure.7
Over the next several days, inorganic solutes (eg, potassium and sodium salts) and various organic solutes are transported out of the cells. In patients in whom this process has had time to occur, treatment of hyponatremia with hypertonic fluids raises the plasma osmolality faster than the cells can recapture the previously transported osmoles. In this situation, overly rapid correction can cause excessive loss of intracellular water, resulting in cell shrinkage and osmotic demyelination syndrome. Osmotic demyelination usually presents during treatment of hyponatremia after an initial improvement in mental status, with worsening neurologic function and various neurologic signs, including paresis and ultimately even death.6
In patients with acute-onset hyponatremia (ie, with onset within the past 48 hours), in whom the above cerebral adaptations have not had time to occur completely, rapid correction is unlikely to result in osmotic demyelination.
In view of the serious risk of osmotic demyelination, if the timing of development of hyponatremia cannot be determined, one should assume it is chronic (> 48 hours) and avoid rapid overcorrection (see discussion below on the rate of correction).
On the other hand, patients who have severe neurologic signs or symptoms initially need their serum sodium increased urgently to safer levels, regardless of the timing of onset (see below for suggested approach). Subsequent treatment of hyponatremia—after the serum sodium level has been raised enough to reverse neurologic symptoms—will be influenced by the duration of the hyponatremia, with careful avoidance of overly rapid correction, especially in patients with chronic hyponatremia.
Assess the patient’s volume status to determine the proper initial treatment
In patients with hypo-osmolar hyponatremia who do not need urgent therapy with hypertonic saline, the initial treatment is based on clinical and laboratory assessment of extracellular fluid volume status, including spot urine sodium measurement (Table 3).3 This will be discussed further below.
Check urine osmolality to assess for hyponatremic states in which urinary dilution is intact
Measuring urine osmolality is useful in ascertaining whether hyponatremic patients are making appropriately dilute urine (< 100 mOsm/kg). If they are, the cause of the hyponatremia may be excessive water intake, a reset osmostat, or low solute intake. In addition, patients with hypovolemic hyponatremia may have appropriately dilute urine soon after treatment with isotonic intravenous fluids.
The serum sodium concentration often returns to normal if the underlying cause is eliminated (eg, if excessive fluid intake is stopped). If there are no serious signs or symptoms, this can usually be accomplished without additional therapy with intravenous fluids or medications, thereby avoiding the risk of overcorrection.
Search for causes of rapidly correctable hyponatremia
If hyponatremia is due to one of several important underlying causes, it may reverse rapidly once the underlying cause has been eliminated (Table 4). Examples: restricting water intake in patients with psychogenic polydipsia, discontinuing thiazide diuretics, replenishing depleted fluid volume, stopping desmopressin (DDAVP), and giving glucocorticoid replacement to those who are glucocorticoid-deficient.
TREATING HYPONATREMIC PATIENTS WITH SERIOUS SIGNS OR SYMPTOMS
Patients with hypo-osmolar hyponatremia and serious signs or symptoms of cerebral edema (lethargy, respiratory depression, seizures) need rapid initial correction of the serum sodium level, as this is a true medical emergency.
Certain patients are at greater risk of developing cerebral edema from hyponatremia (Table 2).
On the other hand, patients with chronic hyponatremia are very unlikely to present with signs or symptoms of cerebral edema. In fact, in a patient with chronic hyponatremia, care must be taken to avoid overcorrection beyond that needed to reverse severe signs and symptoms. In the rare case in which a patient with chronic hyponatremia presents with signs or symptoms of cerebral edema, the hypertonic saline infusion must be stopped as soon as the signs or symptoms have resolved. Further rapid changes in serum sodium must be avoided.
During correction of hyponatremia, some patients are at particularly high risk of osmotic demyelination syndrome secondary to underlying abnormalities in cerebral osmotic regulation. These include patients with alcoholism, malnutrition, hypokalemia, and burns, and elderly women on thiazide diuretics.8 These patients should be monitored vigilantly for overly rapid correction during treatment.
INITIAL TREATMENT: REVERSE CEREBRAL EDEMA WITH 3% SALINE
The goal of the initial, rapid phase of correction is to reverse cerebral edema.
Patients with serious signs or symptoms should receive hypertonic (3%) saline at a rate of about 1 mL/kg/hour for the first several hours.8 Those with concomitant hypervolemia (as in congestive heart failure) or underlying cardiovascular disease should also receive a loop diuretic to aid in free-water excretion and to prevent volume overload from the saline infusion. This regimen usually raises the serum sodium concentration enough (usually by about 1 mmol/L/hour) to decrease cerebral edema and improve symptoms.
In patients having active seizures or showing signs of brain herniation, 3% saline can be given initially at a higher rate of about 2 to 3 mL/kg/hour over the first few hours. An alternative approach is an initial 50-mL bolus of 3% saline and an additional 200 mL given over the subsequent 4 to 6 hours.9
No study has compared the efficacy and safety of these approaches, and clinicians should always monitor extracellular fluid volume status, neurologic status, and serum sodium levels closely in any patient treated with hypertonic saline.
After severe signs and symptoms have resolved, 3% saline is promptly discontinued and appropriate therapy is initiated based on the patient’s volume status and underlying cause of hyponatremia (see discussion below).
NEXT, FIND THE APPROPRIATE RATE OF CORRECTION
After the initial serious signs or symptoms have been addressed with hypertonic saline, management should focus on limiting the rate of correction in patients with chronic hyponatremia or hyponatremia of unknown duration.
Animal studies and retrospective human studies have suggested certain guidelines on the appropriate pace and magnitude of correction during treatment of hyponatremia to avoid osmotic demyelination syndrome.2
Clinicians must not attempt to correct the serum sodium to “normal” values. Although patients with acute hyponatremia may tolerate complete correction, there is little evidence that raising the serum sodium concentration acutely by more than 5 to 8 mmol/L is advantageous. Therefore, correction should be judicious in all patients.
Appropriate rates of correction
A recent expert consensus panel suggested that the serum sodium level be raised by no more than 10 to 12 mmol/L during the first 24 hours of treatment, and by less than 18 mmol/L over 48 hours.2
Patients with chronic hyponatremia and signs or symptoms of cerebral edema should have their sodium level raised at an even slower rate—some recommend less than 10 mmol/L in the first 24 hours.10 Aggressive initial correction at the rate of 1.5 to 2 mmol/hour for the first 3 to 4 hours with 3% saline is indicated until serious symptoms (seizure, obtundation) resolve, but correction beyond 10 to 12 mmol/L in the first 24 hours should be avoided. Hypertonic saline therapy should usually be discontinued well before the serum sodium level has risen this much, to avoid a continuing rise in the sodium level after the infusion has stopped.
While hypertonic saline is being infused, serum sodium levels should be checked every 1 to 2 hours. In a study in 56 patients with severe hyponatremia (serum sodium ≤ 105 mmol/L),11 no neurologic complications were observed in patients with chronic hyponatremia whose serum sodium was corrected by less than 12 mmol/L in 24 hours or by less than 18 mmol/L in 48 hours or in whom the average rate of correction to a serum sodium of 120 mmol/L was less than or equal to 0.55 mmol/L per hour.
If the serum sodium concentration has been overcorrected
Desmopressin is effective in preventing and reversing inadvertent overcorrection of hyponatremia. 12 In one study, desmopressin lowered the sodium concentration by 2 to 9 mmol/L in 14 of 20 patients. None of the patients developed any serious adverse consequences.
In addition, intravenous water (dextrose 5%) can be given alone or in combination with desmopressin to prevent or reverse an excessive increase in serum sodium.13 Such therapy may be considered in patients who continue to excrete hypotonic urine and have already reached a serum sodium concentration that meets or exceeds the recommended rate or magnitude of change.
Formulas for estimating the rate of correction
Various formulas have been devised for estimating the change in serum sodium concentration during treatment of hyponatremia.14
The Adrogué-Madias formula, one of the most commonly used, gives an estimate of how much the serum sodium concentration will rise when 1 L of various intravenous fluids is given (Table 5).15 This formula also accounts for the increase in serum sodium that takes place during concomitant correction of hypokalemia with potassium. Recently, however, a retrospective study16 found that this formula underestimated the change in serum sodium in 23 (74.2%) of 31 patients with hyponatremia treated with hypertonic saline.
An alternative is the Barsoum-Levine equation, which takes into account ongoing urinary losses. Although it is more cumbersome to calculate, it may be more precise.17
Alternatively, in patients without hypovolemia, the clinician can calculate the amount of urinary excretion of free water required to achieve a specific target serum sodium and then measure hourly urinary water excretion during aquaresis induced by furosemide (Lasix).8 Although more physiologic, this method can be clinically cumbersome, requiring timely handling of urine specimens, accurate recording of urine output, and rapid reporting of laboratory results.
Ultimately, these methods serve only as estimates of the change in serum sodium and do not replace careful monitoring of electrolytes (every 1 to 2 hours during acute therapy) and fastidious assessment for clinical signs or symptoms of osmotic demyelination syndrome.
PATIENTS WITH HYPONATREMIA AND NO SERIOUS SIGNS OR SYMPTOMS
General approach
Hyponatremic patients without serious signs or symptoms of cerebral edema do not require urgent therapy to raise the serum sodium.
Patients with chronic asymptomatic hyponatremia are commonly encountered in clinical practice. As a result of cerebral adaptation, they can appear to have no symptoms despite serum sodium levels as low as 115 to 120 mmol/L. However, even if they have no serious signs or symptoms of cerebral edema, some patients may complain of fatigue, lethargy, nausea, gait abnormalities, and muscle cramps and have evidence of milder forms of neurocognitive impairment.18
In a recent case-control study,18 elderly patients with chronic hyponatremia (mean serum sodium concentration 126 ± 5 mmol/L) were more likely to present to the hospital with falls compared with age-matched controls. Further analysis suggested these patients had marked impairments in gait and attention, which improved in some as the serum sodium increased.
Another recent study19 reported that mild hyponatremia (mean serum sodium concentration 132 mmol/L) was independently associated with the risk of fracture, even after adjustment for known osteoporotic risk factors.
Even when there is no need for acute therapy to raise the serum sodium level, the clinician should scrutinize the medical regimen and available clinical data to rule out reversible causes of water excess. These may include ongoing administration of hypotonic fluids (eg, parenteral nutrition or dextrose 5% to “keep the vein open”) or of medications that cause inappropriate release of ADH (eg, selective serotonin reuptake inhibitors) or that impair water excretion (eg, nonsteroidal anti-inflammatory drugs). The clinician should also search for an underlying diagnosis that predisposes to water retention, such as hypothyroidism, adrenal insufficiency, congestive heart failure, or hepatic or renal failure. If hyponatremia is due to endocrine disease, correction of hypothyroidism or adrenal insufficiency should result in water excretion and improvement in the serum sodium.
If the cause of the hyponatremia is not immediately apparent, treatment can be started on the basis of assessment of the patient’s extracellular fluid volume status using clinical examination and supplementary laboratory data such as the serum uric acid concentration and urinary sodium concentration.3Table 3 outlines general treatment options for hypoosmolar hyponatremia according to extracellular fluid volume status.
Of note, physical examination alone has poor sensitivity and specificity in assessing extracellular fluid volume status in patients with hyponatremia.20,21 This highlights the importance of spot measurements of urine sodium and serum uric acid and, when appropriate, isotonic intravenous saline challenge to detect occult hypovolemia.
In general, patients with euvolemia are treated with fluid restriction, and patients with hypovolemia are given isotonic saline. Patients with hypervolemia can be difficult to treat, but in general they are prescribed both sodium and fluid restriction. Loop diuretics can be given to promote excretion of water and sodium. Thiazide diuretics are avoided, as they impair urinary dilution and worsen hyponatremia. Attempts should be made to optimize the treatment of the underlying hypervolemic disorder (congestive heart failure, cirrhosis, advanced renal failure). Vasopressin receptor antagonists can also be used in selected cases of hypervolemic or euvolemic hyponatremia (see discussion below).
How to prescribe fluid restriction rationally
Ideally, patients should not ingest any more fluid than they can excrete in urine and insensible losses—otherwise, the serum sodium can continue to decrease.
Water excretion can be estimated from solute intake and urine osmolarity. In theory, a 70-kg person with a typical daily solute intake of about 10 mOsm/kg and intact urinary dilution to a urine osmolarity of 50 mOsm/L can excrete up to 14 L of urine (700 mOsm/50 mOsm/L) per day. However, a patient with the syndrome of inappropriate ADH secretion (SIADH) and a fixed urine osmolality of 700 mOsm/kg would excrete a similar solute load in only 1 L of urine. Thus, any fluid intake in excess of this volume could worsen hyponatremia.
To excrete free water, urinary sodium plus urinary potassium must be less than the serum sodium concentration. In this regard, the necessary degree of fluid restriction can also be estimated made on the basis of the patient’s urinary electrolytes.22
Increased solute intake to augment water excretion
In patients without hypervolemia, solute intake can be increased to augment water excretion. 22 This can be achieved with salt tablets or oral urea. Although urea can be effective, it is not commonly used because it is not available the United States and it has poor gastrointestinal tolerability. In patients whose nutritional intake is limited and who continue to ingest fluids (such as, for example, an elderly patient subsisting on tea and toast) every effort should be made to increase solute intake with high-protein foods or supplements.
DRUGS TO INHIBIT VASOPRESSIN
Unfortunately, patients often do not adhere to these strategies, as fluid restriction and unpalatable salt tablets or urea can become too burdensome. In such instances, pharmacologic inhibition of vasopressin-mediated water reabsorption can be considered using the following agents.
Demeclocycline (Declomycin) and lithium inhibit the kidney’s response to vasopressin. Because lithium may be nephrotoxic and has unwanted effects on the central nervous system, demeclocycline has become the preferred agent. Given in doses of 300 to 600 mg twice daily, demeclocycline promotes free water excretion, but often takes 1 to 2 weeks of therapy to begin working.
Renal failure due to demeclocycline has been reported in patients with concomitant liver disease.23 Demeclocycline can also cause photosensitivity and is contraindicated in children and pregnant women due to abnormalities in bone and enamel formation. In addition, it can be expensive and may not be covered fully by some prescription plans.
Vasopressin receptor antagonists (‘vaptans’)
ADH, also called vasopressin, interacts with various receptor subtypes, including V1a (causing vasoconstriction, platelet aggregation, inotropic stimulation, myocardial protein synthesis), V1b (causing secretion of adrenocorticotropic hormone), and V2 (causing water reabsorption and release of von Willebrand factor and factor VIII).
Drugs that block V2 receptors in the renal tubule increase water excretion, making them attractive as therapy for some hyponatremic states (Table 6).24,25 These drugs exert their aquaretic effect by causing a decrease in transcription and insertion of aquaporin-2 channels (“water pores”) into the apical collecting duct membrane. As a result, the water permeability of the collecting duct is decreased even in the presence of circulating ADH.
Conivaptan (Vaprisol) is a combined V1a-V2 antagonist that has been approved for the treatment of euvolemic and hypervolemic hyponatremia. Conivaptan inhibits the cytochrome P450 3A4 system and thus may interact with other drugs; therefore, its use has been limited to no more than 4 days of intravenous administration in the hospital setting. The recommended dosage is an initial 20-mg infusion over 30 minutes, followed by continuous infusions of 20 to 40 mg/day. Dosing adjustments in renal and hepatic impairment have not been well defined.
Tolvaptan (Samsca) is an oral selective V2 antagonist that has been studied in patients with euvolemic and hypervolemic hyponatremia. 26 Studies have included patients with congestive heart failure, cirrhosis, and SIADH. Although tolvaptan has not been shown to reduce rates of rehospitalization or death in congestive heart failure, it improves serum sodium, overall fluid balance, and congestive symptoms.27 Tolvaptan has recently been approved for the treatment of euvolemic and hypervolemic hyponatremia.
A recent study has confirmed the longterm efficacy of tolvaptan in 111 patients over a mean duration of treatment greater than 700 days.28 While the clinical benefits of chronic tolvaptan therapy have yet to be clearly demonstrated, this study shows that tolvaptan therapy can result in a sustained improvement in serum sodium concentration without an unacceptable increase in adverse events.29
Lixivaptan (VPA-985), another oral selective V2 receptor antagonist, is being studied in patients with euvolemic and hypervolemic hyponatremia.
Current role of vasopressin antagonists
Current studies of vasopressin antagonists in the treatment of hyponatremia are promising, though definite recommendations are needed to ensure slow, careful correction of hyponatremia. Most studies suggest that these agents provide slow, reliable increases in serum sodium. In one large study of patients with congestive heart failure, serum sodium rose by more than 12 mmol/L in 24 hours in fewer than 2% of patients.26
Notably, no cases of osmotic demyelination syndrome have been reported in these studies. However, it should be noted that therapy was started in the hospital with close monitoring of serum sodium levels and discontinuation of fluid restriction; the incidence of overly rapid correction of sodium may be higher outside of carefully done clinical studies. Clinicians should adopt monitoring strategies similar to those used in these rigorous studies.
At present, there is little experience with vasopressin antagonists in hyponatremic patients with serious signs or symptoms of cerebral edema, and most clinicians still view 3% saline as the gold standard for these patients.
Vasopressin antagonists should not be used in patients with hypovolemic hyponatremia, due to concerns about V1a blockade causing hypotension and about V2 blockade producing water excretion and a worsening of the volume-depleted state.
Recent clinical trials have reported that patients often experience increased thirst while taking these agents. This highlights the need to monitor serum sodium during treatment.
These agents are expensive. Tolvaptan costs about $250 per tablet; conivaptan, which is administered intravenously, may cost a little more per treatment course.
THERAPY IN SPECIFIC DISEASE STATES
Patients with hyponatremia and cirrhosis
The focus of treatment remains water and salt restriction and judicious use of loop diuretics and aldosterone antagonists such as spironolactone (Aldactone).
Tolvaptan has been effective at raising the serum sodium level in patients with cirrhosis, 26 while conivaptan should be avoided at present because of vasodilation from V1a receptor antagonism and its potential effects on systemic hemodynamics and risk of variceal bleeding.30
As the severity of cirrhosis increases, the only effective treatment of hyponatremia is liver transplantation.
Patients with SIADH
In most cases, water restriction is the mainstay of therapy. Adequate nutritional intake should also be stressed so that enough solute is available for ongoing water excretion. Although fluid restriction is usually effective, many patients cannot adhere to the level of restriction required.
In cases in which fluid restriction is not effective on its own, demeclocyline can be used to antagonize ADH action and increase water excretion. Sodium tablets and loop diuretics can also be used, taking care to avoid hypovolemia from diuretic-induced sodium losses. The use of tolvaptan in patients with SIADH has resulted in short-term increases in serum sodium.26 A recent study has suggested that this effect can be sustained with longer-term treatment,28 but further studies are needed to show a complementary clinical benefit (eg, improved neurocognition) to guide the use of these costly agents in clinical practice.
Patients with diuretic-induced hyponatremia
Thiazide diuretics should be discontinued and hypovolemia and hypokalemia should be corrected with isotonic saline and potassium supplementation. As the hypokalemia is corrected and the diuretic effect and hypovolemic stimulus to ADH dissipates, water excretion can increase rapidly, resulting in a brisk change in serum sodium.
Serum sodium levels should be closely monitored during therapy to avoid overcorrection. For this reason, use of hypertonic saline should generally be avoided. Hypotonic fluid— eg, half-normal (0.45%) or quarter-normal (0.22%) saline or even desmopressin—may become necessary in the later stages of therapy to avoid overly rapid correction.
Patients with exercise-associated hyponatremia
Patients at highest risk of exercise-associated hyponatremia include those who drink too much fluid during a long-distance race, who have low body weight, who are female, who exercise longer than 4 hours, and who use nonsteroidal anti-inflammatory drugs.31 The cause of hyponatremia is likely multifactorial, with excessive water intake coupled with sodium losses and impaired renal excretion of water due to ADH action and impaired renal dilution. To prevent exercise-associated hyponatremia, fluid intake should be limited to 400 to 800 mL/hour, with the higher end recommended for larger athletes and hotter climates.
Consensus recommendations suggest that most patients with mild hyponatremia (serum sodium 130 to 135 mmol/L) should be treated with fluid restriction and clinical observation, as spontaneous water diuresis leads to improvement in the serum sodium level. Importantly, the reflex to provide isotonic saline infusions should be avoided unless clear signs of volume depletion are present. Intravenous saline has the potential to worsen hyponatremia in the presence of ADH. In addition, some athletes will have retained water in the gastrointestinal tract that may be mobilized after the race, resulting in worsening of hyponatremia.32
In athletes with severe hyponatremia (serum sodium < 120 mmol/L) or symptomatic exercise-associated hyponatremia (lethargy, respiratory depression, seizures), hypertonic saline is the treatment of choice. One protocol suggests giving 100 mL of 3% saline over 10 minutes in the field, followed by prompt transportation to hospital.33
SUMMARY POINTS
Hyponatremia is a common electrolyte disorder that in its most severe form requires urgent therapy with hypertonic saline to correct cerebral edema.
In patients without serious signs or symptoms of cerebral edema, recent observations suggest there may be clinically important symptomatology relating to mild neurocognitive dysfunction and an association with risk of bone fracture.
Multiple treatment strategies are available according to the underlying extracellular fluid volume status and cause of hyponatremia. These include fluid and sodium restriction and augmentation of urinary water excretion with various nutritional and pharmacologic strategies. The most novel therapy includes antagonism of the vasopressin V2 receptor with a class of aquaretic agents known as vaptans.
There can be serious neurologic injury associated with overly rapid correction of chronic hyponatremia or undercorrection of acute symptomatic hyponatremia.
Clinicians must be familiar with the details of each of the treatments and have an appreciation of the importance of careful monitoring during treatment.
Hyponatremia, defined as a serum sodium concentration below 135 mmol/L, is one of the most frequently encountered electrolyte disorders. In 1981, Flear et al1 reported that 15% of their hospitalized patients had plasma sodium concentrations lower than 134 mmol/L, the cutoff they were using at that time.
Hyponatremia is sometimes merely a laboratory artifact or a result of improper blood collection. If real, it can be due to excessive water intake or, most often, the inability of the kidney to excrete water coupled with continued water intake. Patients with significant underlying cardiac, hepatic, or renal dysfunction are at greatest risk of developing hyponatremia, secondary to the nonosmotic release of antidiuretic hormone (ADH). Others at risk include postoperative patients (especially menstruating women), older patients on thiazide diuretics, patients with malignant or psychiatric illness, and endurance athletes.
In this article, we review the treatment of acute and chronic hyponatremia, emphasizing the importance of basing the therapy on the severity of symptoms and taking care not to raise the serum sodium level too rapidly, which can cause neurologic dysfunction.
Guidelines for managing hyponatremia2 are based mostly on retrospective studies and expert opinion, since few prospective studies have been done. Despite the paucity of evidence-based recommendations, we will attempt to incorporate findings from important human and animal studies and consensus guidelines from expert panels. We will focus initially on the critical diagnostic considerations necessary to initiate treatment.
SYMPTOMATIC VS ASYMPTOMATIC
Subsequent sections will address therapeutic approaches in two clinical settings:
Symptomatic hyponatremia, ie, with severe signs or symptoms of cerebral edema—a medical emergency; and
Asymptomatic hyponatremia, ie, without serious signs or symptoms of cerebral edema.
KEY DIAGNOSTIC STEPS WHEN STARTING TREATMENT
The treatment of hyponatremia begins by confirming a truly hypo-osmolar state, assessing its clinical significance, and determining its cause (Table 1).
The clinical and laboratory evaluations form the foundation of a proper approach to any patient with hyponatremia. The rationale behind making several important diagnostic distinctions will be discussed here briefly and then expanded on in the remaining text. The reader is referred to another review on the diagnostic evaluation of hyponatremia.3
Confirm that the patient truly has hypo-osmolar hyponatremia
The serum osmolality should be measured to confirm that it is low (ie, < 275 mOsm/kg). In addition, the arterial serum sodium concentration can be measured using a blood gas device if pseudohyponatremia (see below) is suspected. This method uses direct potentiometry and bypasses the dilutional step in the processing of venous samples.4
Rationale. The clinical consequences of hyponatremia are due to water moving from hypo-osmolar extracellular fluid into the relatively hyperosmolar interior of the cell. This water movement can cause progressive cerebral edema, resulting in a spectrum of signs and symptoms from headache and ataxia to seizures and coma. But significant fluid shifts and cerebral edema occur only if the extracellular fluid is hypo-osmolar relative to the intracellular fluid.
In fact, hyponatremia can occur in several situations in which the extracellular fluid is not hypo-osmolar. An increase in effective plasma osmoles (substances in the extracellular fluid that do not readily move across the plasma membrane) can cause water to move out of cells, resulting in translocational hyponatremia. This may be seen in hyperglycemia or when mannitol or contrast dye has been given. In these situations, the plasma is either isotonic or even hypertonic to the intracellular fluid, resulting in no movement of water into the cells and therefore no clinical consequences relating to the hyponatremia. Importantly, no therapy is required for the hyponatremia.
Other situations in which hyponatremia is present but not associated with true hypotonicity include states of excess protein or lipid in the blood (pseudohyponatremia). Also, if an infusion of hypotonic fluid is running, clinicians must be sure that blood samples are not drawn proximally in the same vein.
Are there significant signs or symptoms of cerebral edema?
Hyponatremia can cause brain swelling within the confined space of the skull as water shifts from the extracellular fluid into the cells. Depending on underlying risk factors (Table 2)5 and the severity and duration of hyponatremia (see below), this may result in signs or symptoms of cerebral edema, including visual changes, focal neurologic changes, encephalopathy, respiratory depression, and seizures. Ultimately, brain herniation can occur.
Patients need to be assessed quickly because those with serious neurologic signs or symptoms thought to be related to hyponatremia require urgent treatment with hypertonic saline to increase the serum sodium concentration, regardless of the underlying volume status, the cause of hyponatremia, or the time of onset.
Determine the duration of hyponatremia
One should try to ascertain when the hyponatremia started, as its duration is important in determining the proper pace of correction.
Figure 1.The brain begins to adapt to hyponatremia within minutes, and the cerebral adaptation is maximal within 2 to 3 days (Figure 1).6
At the onset of hyponatremia, water moves from the extracellular fluid into cells, pulled in by osmosis. The brain can decrease the net amount of water entering into the neurons (and thus regulate its volume) by increasing the flow of water from the interstitium into the cerebrospinal fluid via increased interstitial hydraulic pressure.7
Over the next several days, inorganic solutes (eg, potassium and sodium salts) and various organic solutes are transported out of the cells. In patients in whom this process has had time to occur, treatment of hyponatremia with hypertonic fluids raises the plasma osmolality faster than the cells can recapture the previously transported osmoles. In this situation, overly rapid correction can cause excessive loss of intracellular water, resulting in cell shrinkage and osmotic demyelination syndrome. Osmotic demyelination usually presents during treatment of hyponatremia after an initial improvement in mental status, with worsening neurologic function and various neurologic signs, including paresis and ultimately even death.6
In patients with acute-onset hyponatremia (ie, with onset within the past 48 hours), in whom the above cerebral adaptations have not had time to occur completely, rapid correction is unlikely to result in osmotic demyelination.
In view of the serious risk of osmotic demyelination, if the timing of development of hyponatremia cannot be determined, one should assume it is chronic (> 48 hours) and avoid rapid overcorrection (see discussion below on the rate of correction).
On the other hand, patients who have severe neurologic signs or symptoms initially need their serum sodium increased urgently to safer levels, regardless of the timing of onset (see below for suggested approach). Subsequent treatment of hyponatremia—after the serum sodium level has been raised enough to reverse neurologic symptoms—will be influenced by the duration of the hyponatremia, with careful avoidance of overly rapid correction, especially in patients with chronic hyponatremia.
Assess the patient’s volume status to determine the proper initial treatment
In patients with hypo-osmolar hyponatremia who do not need urgent therapy with hypertonic saline, the initial treatment is based on clinical and laboratory assessment of extracellular fluid volume status, including spot urine sodium measurement (Table 3).3 This will be discussed further below.
Check urine osmolality to assess for hyponatremic states in which urinary dilution is intact
Measuring urine osmolality is useful in ascertaining whether hyponatremic patients are making appropriately dilute urine (< 100 mOsm/kg). If they are, the cause of the hyponatremia may be excessive water intake, a reset osmostat, or low solute intake. In addition, patients with hypovolemic hyponatremia may have appropriately dilute urine soon after treatment with isotonic intravenous fluids.
The serum sodium concentration often returns to normal if the underlying cause is eliminated (eg, if excessive fluid intake is stopped). If there are no serious signs or symptoms, this can usually be accomplished without additional therapy with intravenous fluids or medications, thereby avoiding the risk of overcorrection.
Search for causes of rapidly correctable hyponatremia
If hyponatremia is due to one of several important underlying causes, it may reverse rapidly once the underlying cause has been eliminated (Table 4). Examples: restricting water intake in patients with psychogenic polydipsia, discontinuing thiazide diuretics, replenishing depleted fluid volume, stopping desmopressin (DDAVP), and giving glucocorticoid replacement to those who are glucocorticoid-deficient.
TREATING HYPONATREMIC PATIENTS WITH SERIOUS SIGNS OR SYMPTOMS
Patients with hypo-osmolar hyponatremia and serious signs or symptoms of cerebral edema (lethargy, respiratory depression, seizures) need rapid initial correction of the serum sodium level, as this is a true medical emergency.
Certain patients are at greater risk of developing cerebral edema from hyponatremia (Table 2).
On the other hand, patients with chronic hyponatremia are very unlikely to present with signs or symptoms of cerebral edema. In fact, in a patient with chronic hyponatremia, care must be taken to avoid overcorrection beyond that needed to reverse severe signs and symptoms. In the rare case in which a patient with chronic hyponatremia presents with signs or symptoms of cerebral edema, the hypertonic saline infusion must be stopped as soon as the signs or symptoms have resolved. Further rapid changes in serum sodium must be avoided.
During correction of hyponatremia, some patients are at particularly high risk of osmotic demyelination syndrome secondary to underlying abnormalities in cerebral osmotic regulation. These include patients with alcoholism, malnutrition, hypokalemia, and burns, and elderly women on thiazide diuretics.8 These patients should be monitored vigilantly for overly rapid correction during treatment.
INITIAL TREATMENT: REVERSE CEREBRAL EDEMA WITH 3% SALINE
The goal of the initial, rapid phase of correction is to reverse cerebral edema.
Patients with serious signs or symptoms should receive hypertonic (3%) saline at a rate of about 1 mL/kg/hour for the first several hours.8 Those with concomitant hypervolemia (as in congestive heart failure) or underlying cardiovascular disease should also receive a loop diuretic to aid in free-water excretion and to prevent volume overload from the saline infusion. This regimen usually raises the serum sodium concentration enough (usually by about 1 mmol/L/hour) to decrease cerebral edema and improve symptoms.
In patients having active seizures or showing signs of brain herniation, 3% saline can be given initially at a higher rate of about 2 to 3 mL/kg/hour over the first few hours. An alternative approach is an initial 50-mL bolus of 3% saline and an additional 200 mL given over the subsequent 4 to 6 hours.9
No study has compared the efficacy and safety of these approaches, and clinicians should always monitor extracellular fluid volume status, neurologic status, and serum sodium levels closely in any patient treated with hypertonic saline.
After severe signs and symptoms have resolved, 3% saline is promptly discontinued and appropriate therapy is initiated based on the patient’s volume status and underlying cause of hyponatremia (see discussion below).
NEXT, FIND THE APPROPRIATE RATE OF CORRECTION
After the initial serious signs or symptoms have been addressed with hypertonic saline, management should focus on limiting the rate of correction in patients with chronic hyponatremia or hyponatremia of unknown duration.
Animal studies and retrospective human studies have suggested certain guidelines on the appropriate pace and magnitude of correction during treatment of hyponatremia to avoid osmotic demyelination syndrome.2
Clinicians must not attempt to correct the serum sodium to “normal” values. Although patients with acute hyponatremia may tolerate complete correction, there is little evidence that raising the serum sodium concentration acutely by more than 5 to 8 mmol/L is advantageous. Therefore, correction should be judicious in all patients.
Appropriate rates of correction
A recent expert consensus panel suggested that the serum sodium level be raised by no more than 10 to 12 mmol/L during the first 24 hours of treatment, and by less than 18 mmol/L over 48 hours.2
Patients with chronic hyponatremia and signs or symptoms of cerebral edema should have their sodium level raised at an even slower rate—some recommend less than 10 mmol/L in the first 24 hours.10 Aggressive initial correction at the rate of 1.5 to 2 mmol/hour for the first 3 to 4 hours with 3% saline is indicated until serious symptoms (seizure, obtundation) resolve, but correction beyond 10 to 12 mmol/L in the first 24 hours should be avoided. Hypertonic saline therapy should usually be discontinued well before the serum sodium level has risen this much, to avoid a continuing rise in the sodium level after the infusion has stopped.
While hypertonic saline is being infused, serum sodium levels should be checked every 1 to 2 hours. In a study in 56 patients with severe hyponatremia (serum sodium ≤ 105 mmol/L),11 no neurologic complications were observed in patients with chronic hyponatremia whose serum sodium was corrected by less than 12 mmol/L in 24 hours or by less than 18 mmol/L in 48 hours or in whom the average rate of correction to a serum sodium of 120 mmol/L was less than or equal to 0.55 mmol/L per hour.
If the serum sodium concentration has been overcorrected
Desmopressin is effective in preventing and reversing inadvertent overcorrection of hyponatremia. 12 In one study, desmopressin lowered the sodium concentration by 2 to 9 mmol/L in 14 of 20 patients. None of the patients developed any serious adverse consequences.
In addition, intravenous water (dextrose 5%) can be given alone or in combination with desmopressin to prevent or reverse an excessive increase in serum sodium.13 Such therapy may be considered in patients who continue to excrete hypotonic urine and have already reached a serum sodium concentration that meets or exceeds the recommended rate or magnitude of change.
Formulas for estimating the rate of correction
Various formulas have been devised for estimating the change in serum sodium concentration during treatment of hyponatremia.14
The Adrogué-Madias formula, one of the most commonly used, gives an estimate of how much the serum sodium concentration will rise when 1 L of various intravenous fluids is given (Table 5).15 This formula also accounts for the increase in serum sodium that takes place during concomitant correction of hypokalemia with potassium. Recently, however, a retrospective study16 found that this formula underestimated the change in serum sodium in 23 (74.2%) of 31 patients with hyponatremia treated with hypertonic saline.
An alternative is the Barsoum-Levine equation, which takes into account ongoing urinary losses. Although it is more cumbersome to calculate, it may be more precise.17
Alternatively, in patients without hypovolemia, the clinician can calculate the amount of urinary excretion of free water required to achieve a specific target serum sodium and then measure hourly urinary water excretion during aquaresis induced by furosemide (Lasix).8 Although more physiologic, this method can be clinically cumbersome, requiring timely handling of urine specimens, accurate recording of urine output, and rapid reporting of laboratory results.
Ultimately, these methods serve only as estimates of the change in serum sodium and do not replace careful monitoring of electrolytes (every 1 to 2 hours during acute therapy) and fastidious assessment for clinical signs or symptoms of osmotic demyelination syndrome.
PATIENTS WITH HYPONATREMIA AND NO SERIOUS SIGNS OR SYMPTOMS
General approach
Hyponatremic patients without serious signs or symptoms of cerebral edema do not require urgent therapy to raise the serum sodium.
Patients with chronic asymptomatic hyponatremia are commonly encountered in clinical practice. As a result of cerebral adaptation, they can appear to have no symptoms despite serum sodium levels as low as 115 to 120 mmol/L. However, even if they have no serious signs or symptoms of cerebral edema, some patients may complain of fatigue, lethargy, nausea, gait abnormalities, and muscle cramps and have evidence of milder forms of neurocognitive impairment.18
In a recent case-control study,18 elderly patients with chronic hyponatremia (mean serum sodium concentration 126 ± 5 mmol/L) were more likely to present to the hospital with falls compared with age-matched controls. Further analysis suggested these patients had marked impairments in gait and attention, which improved in some as the serum sodium increased.
Another recent study19 reported that mild hyponatremia (mean serum sodium concentration 132 mmol/L) was independently associated with the risk of fracture, even after adjustment for known osteoporotic risk factors.
Even when there is no need for acute therapy to raise the serum sodium level, the clinician should scrutinize the medical regimen and available clinical data to rule out reversible causes of water excess. These may include ongoing administration of hypotonic fluids (eg, parenteral nutrition or dextrose 5% to “keep the vein open”) or of medications that cause inappropriate release of ADH (eg, selective serotonin reuptake inhibitors) or that impair water excretion (eg, nonsteroidal anti-inflammatory drugs). The clinician should also search for an underlying diagnosis that predisposes to water retention, such as hypothyroidism, adrenal insufficiency, congestive heart failure, or hepatic or renal failure. If hyponatremia is due to endocrine disease, correction of hypothyroidism or adrenal insufficiency should result in water excretion and improvement in the serum sodium.
If the cause of the hyponatremia is not immediately apparent, treatment can be started on the basis of assessment of the patient’s extracellular fluid volume status using clinical examination and supplementary laboratory data such as the serum uric acid concentration and urinary sodium concentration.3Table 3 outlines general treatment options for hypoosmolar hyponatremia according to extracellular fluid volume status.
Of note, physical examination alone has poor sensitivity and specificity in assessing extracellular fluid volume status in patients with hyponatremia.20,21 This highlights the importance of spot measurements of urine sodium and serum uric acid and, when appropriate, isotonic intravenous saline challenge to detect occult hypovolemia.
In general, patients with euvolemia are treated with fluid restriction, and patients with hypovolemia are given isotonic saline. Patients with hypervolemia can be difficult to treat, but in general they are prescribed both sodium and fluid restriction. Loop diuretics can be given to promote excretion of water and sodium. Thiazide diuretics are avoided, as they impair urinary dilution and worsen hyponatremia. Attempts should be made to optimize the treatment of the underlying hypervolemic disorder (congestive heart failure, cirrhosis, advanced renal failure). Vasopressin receptor antagonists can also be used in selected cases of hypervolemic or euvolemic hyponatremia (see discussion below).
How to prescribe fluid restriction rationally
Ideally, patients should not ingest any more fluid than they can excrete in urine and insensible losses—otherwise, the serum sodium can continue to decrease.
Water excretion can be estimated from solute intake and urine osmolarity. In theory, a 70-kg person with a typical daily solute intake of about 10 mOsm/kg and intact urinary dilution to a urine osmolarity of 50 mOsm/L can excrete up to 14 L of urine (700 mOsm/50 mOsm/L) per day. However, a patient with the syndrome of inappropriate ADH secretion (SIADH) and a fixed urine osmolality of 700 mOsm/kg would excrete a similar solute load in only 1 L of urine. Thus, any fluid intake in excess of this volume could worsen hyponatremia.
To excrete free water, urinary sodium plus urinary potassium must be less than the serum sodium concentration. In this regard, the necessary degree of fluid restriction can also be estimated made on the basis of the patient’s urinary electrolytes.22
Increased solute intake to augment water excretion
In patients without hypervolemia, solute intake can be increased to augment water excretion. 22 This can be achieved with salt tablets or oral urea. Although urea can be effective, it is not commonly used because it is not available the United States and it has poor gastrointestinal tolerability. In patients whose nutritional intake is limited and who continue to ingest fluids (such as, for example, an elderly patient subsisting on tea and toast) every effort should be made to increase solute intake with high-protein foods or supplements.
DRUGS TO INHIBIT VASOPRESSIN
Unfortunately, patients often do not adhere to these strategies, as fluid restriction and unpalatable salt tablets or urea can become too burdensome. In such instances, pharmacologic inhibition of vasopressin-mediated water reabsorption can be considered using the following agents.
Demeclocycline (Declomycin) and lithium inhibit the kidney’s response to vasopressin. Because lithium may be nephrotoxic and has unwanted effects on the central nervous system, demeclocycline has become the preferred agent. Given in doses of 300 to 600 mg twice daily, demeclocycline promotes free water excretion, but often takes 1 to 2 weeks of therapy to begin working.
Renal failure due to demeclocycline has been reported in patients with concomitant liver disease.23 Demeclocycline can also cause photosensitivity and is contraindicated in children and pregnant women due to abnormalities in bone and enamel formation. In addition, it can be expensive and may not be covered fully by some prescription plans.
Vasopressin receptor antagonists (‘vaptans’)
ADH, also called vasopressin, interacts with various receptor subtypes, including V1a (causing vasoconstriction, platelet aggregation, inotropic stimulation, myocardial protein synthesis), V1b (causing secretion of adrenocorticotropic hormone), and V2 (causing water reabsorption and release of von Willebrand factor and factor VIII).
Drugs that block V2 receptors in the renal tubule increase water excretion, making them attractive as therapy for some hyponatremic states (Table 6).24,25 These drugs exert their aquaretic effect by causing a decrease in transcription and insertion of aquaporin-2 channels (“water pores”) into the apical collecting duct membrane. As a result, the water permeability of the collecting duct is decreased even in the presence of circulating ADH.
Conivaptan (Vaprisol) is a combined V1a-V2 antagonist that has been approved for the treatment of euvolemic and hypervolemic hyponatremia. Conivaptan inhibits the cytochrome P450 3A4 system and thus may interact with other drugs; therefore, its use has been limited to no more than 4 days of intravenous administration in the hospital setting. The recommended dosage is an initial 20-mg infusion over 30 minutes, followed by continuous infusions of 20 to 40 mg/day. Dosing adjustments in renal and hepatic impairment have not been well defined.
Tolvaptan (Samsca) is an oral selective V2 antagonist that has been studied in patients with euvolemic and hypervolemic hyponatremia. 26 Studies have included patients with congestive heart failure, cirrhosis, and SIADH. Although tolvaptan has not been shown to reduce rates of rehospitalization or death in congestive heart failure, it improves serum sodium, overall fluid balance, and congestive symptoms.27 Tolvaptan has recently been approved for the treatment of euvolemic and hypervolemic hyponatremia.
A recent study has confirmed the longterm efficacy of tolvaptan in 111 patients over a mean duration of treatment greater than 700 days.28 While the clinical benefits of chronic tolvaptan therapy have yet to be clearly demonstrated, this study shows that tolvaptan therapy can result in a sustained improvement in serum sodium concentration without an unacceptable increase in adverse events.29
Lixivaptan (VPA-985), another oral selective V2 receptor antagonist, is being studied in patients with euvolemic and hypervolemic hyponatremia.
Current role of vasopressin antagonists
Current studies of vasopressin antagonists in the treatment of hyponatremia are promising, though definite recommendations are needed to ensure slow, careful correction of hyponatremia. Most studies suggest that these agents provide slow, reliable increases in serum sodium. In one large study of patients with congestive heart failure, serum sodium rose by more than 12 mmol/L in 24 hours in fewer than 2% of patients.26
Notably, no cases of osmotic demyelination syndrome have been reported in these studies. However, it should be noted that therapy was started in the hospital with close monitoring of serum sodium levels and discontinuation of fluid restriction; the incidence of overly rapid correction of sodium may be higher outside of carefully done clinical studies. Clinicians should adopt monitoring strategies similar to those used in these rigorous studies.
At present, there is little experience with vasopressin antagonists in hyponatremic patients with serious signs or symptoms of cerebral edema, and most clinicians still view 3% saline as the gold standard for these patients.
Vasopressin antagonists should not be used in patients with hypovolemic hyponatremia, due to concerns about V1a blockade causing hypotension and about V2 blockade producing water excretion and a worsening of the volume-depleted state.
Recent clinical trials have reported that patients often experience increased thirst while taking these agents. This highlights the need to monitor serum sodium during treatment.
These agents are expensive. Tolvaptan costs about $250 per tablet; conivaptan, which is administered intravenously, may cost a little more per treatment course.
THERAPY IN SPECIFIC DISEASE STATES
Patients with hyponatremia and cirrhosis
The focus of treatment remains water and salt restriction and judicious use of loop diuretics and aldosterone antagonists such as spironolactone (Aldactone).
Tolvaptan has been effective at raising the serum sodium level in patients with cirrhosis, 26 while conivaptan should be avoided at present because of vasodilation from V1a receptor antagonism and its potential effects on systemic hemodynamics and risk of variceal bleeding.30
As the severity of cirrhosis increases, the only effective treatment of hyponatremia is liver transplantation.
Patients with SIADH
In most cases, water restriction is the mainstay of therapy. Adequate nutritional intake should also be stressed so that enough solute is available for ongoing water excretion. Although fluid restriction is usually effective, many patients cannot adhere to the level of restriction required.
In cases in which fluid restriction is not effective on its own, demeclocyline can be used to antagonize ADH action and increase water excretion. Sodium tablets and loop diuretics can also be used, taking care to avoid hypovolemia from diuretic-induced sodium losses. The use of tolvaptan in patients with SIADH has resulted in short-term increases in serum sodium.26 A recent study has suggested that this effect can be sustained with longer-term treatment,28 but further studies are needed to show a complementary clinical benefit (eg, improved neurocognition) to guide the use of these costly agents in clinical practice.
Patients with diuretic-induced hyponatremia
Thiazide diuretics should be discontinued and hypovolemia and hypokalemia should be corrected with isotonic saline and potassium supplementation. As the hypokalemia is corrected and the diuretic effect and hypovolemic stimulus to ADH dissipates, water excretion can increase rapidly, resulting in a brisk change in serum sodium.
Serum sodium levels should be closely monitored during therapy to avoid overcorrection. For this reason, use of hypertonic saline should generally be avoided. Hypotonic fluid— eg, half-normal (0.45%) or quarter-normal (0.22%) saline or even desmopressin—may become necessary in the later stages of therapy to avoid overly rapid correction.
Patients with exercise-associated hyponatremia
Patients at highest risk of exercise-associated hyponatremia include those who drink too much fluid during a long-distance race, who have low body weight, who are female, who exercise longer than 4 hours, and who use nonsteroidal anti-inflammatory drugs.31 The cause of hyponatremia is likely multifactorial, with excessive water intake coupled with sodium losses and impaired renal excretion of water due to ADH action and impaired renal dilution. To prevent exercise-associated hyponatremia, fluid intake should be limited to 400 to 800 mL/hour, with the higher end recommended for larger athletes and hotter climates.
Consensus recommendations suggest that most patients with mild hyponatremia (serum sodium 130 to 135 mmol/L) should be treated with fluid restriction and clinical observation, as spontaneous water diuresis leads to improvement in the serum sodium level. Importantly, the reflex to provide isotonic saline infusions should be avoided unless clear signs of volume depletion are present. Intravenous saline has the potential to worsen hyponatremia in the presence of ADH. In addition, some athletes will have retained water in the gastrointestinal tract that may be mobilized after the race, resulting in worsening of hyponatremia.32
In athletes with severe hyponatremia (serum sodium < 120 mmol/L) or symptomatic exercise-associated hyponatremia (lethargy, respiratory depression, seizures), hypertonic saline is the treatment of choice. One protocol suggests giving 100 mL of 3% saline over 10 minutes in the field, followed by prompt transportation to hospital.33
SUMMARY POINTS
Hyponatremia is a common electrolyte disorder that in its most severe form requires urgent therapy with hypertonic saline to correct cerebral edema.
In patients without serious signs or symptoms of cerebral edema, recent observations suggest there may be clinically important symptomatology relating to mild neurocognitive dysfunction and an association with risk of bone fracture.
Multiple treatment strategies are available according to the underlying extracellular fluid volume status and cause of hyponatremia. These include fluid and sodium restriction and augmentation of urinary water excretion with various nutritional and pharmacologic strategies. The most novel therapy includes antagonism of the vasopressin V2 receptor with a class of aquaretic agents known as vaptans.
There can be serious neurologic injury associated with overly rapid correction of chronic hyponatremia or undercorrection of acute symptomatic hyponatremia.
Clinicians must be familiar with the details of each of the treatments and have an appreciation of the importance of careful monitoring during treatment.
References
Flear CTG, Gill GV, Burn J. Hyponatremia: mechanisms and management. Lancet1981; 2:26–31.
Freda BJ, Davidson MB, Hall PM. Evaluation of hyponatremia: a little physiology goes a long way. Cleve Clin J Med2004; 71:639–650.
Weisberg LS. Pseudohyponatremia: a reappraisal. Am J Med1989; 86:315–318.
Moritz L, Ayus JC. The pathophysiology and treatment of hyponatraemic encephalopathy: an update. Nephrol Dial Transplant2003; 18:2486–2491.
Widdess-Walsh P, Sabharwal V, Demirjian S, DeGeorgia M. Neurologic effects of hyponatremia and its treatment. Cleve Clin J Med2007; 74:377–383.
Melton JE, Patlak CS, Pettigrew KD, Cserr HF. Volume regulatory loss of Na, Cl, and K from rat brain during acute hyponatremia. Am J Physiol1987; 252:F661–F669.
Lauriat SM, Berl T. The hyponatremic patient: practical focus on therapy. J Am Soc Nephrol1997; 8:1599–1607.
Kokko JP. Symptomatic hyponatremia with hypoxia is a medical emergency. Kidney Int2006; 69:1291–1293.
Ellis SJ. Severe hyponatraemia: complications and treatment. QJM1995; 88:905–909.
Sterns RH, Cappuccio JD, Silver SM, Cohen EP. Neurologic sequelae after treatment of severe hyponatremia: a multicenter perspective. J Am Soc Nephrol1994; 4:1522–1530.
Perianayagam A, Sterns RH, Silver SM, et al. DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia. Clin J Am Soc Nephrol2008; 3:331–336.
Sterns RH, Hix JK. Overcorrection of hyponatremia is a medical emergency. Kidney Int2009; 76:587–589.
Nguyen MK, Kurtz I. Analysis of current formulas used for treatment of the dysnatremias. Clin Exp Nephrol2004; 8:12–16.
Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med2000; 342:1581–1589.
Mohmand HK, Issa D, Ahmad Z, Cappuccio JD, Kouides RW, Sterns RH. Hypertonic saline for hyponatremia: risk of inadvertent overcorrection. Clin J Am Soc Nephrol2007; 2:1110–1117.
Ellison DH, Berl T. Clinical practice. The syndrome of inappropriate antidiuresis. N Engl J Med2007; 356:2064–2072.
Renneboog B, Musch W, Vandemergel X, Manto MU, Decaux G. Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits. Am J Med2006; 119:71.e1–e8.
Kinsella S, Moran S, Sullivan MO, Molloy MG, Eustace JA. Hyponatremia independent of osteoporosis is associated with fracture occurrence. Clin J Am Soc Nephrol2010; 5:275–280.
Chung HM, Kluge R, Schrier RW, Anderson RJ. Clinical assessment of extracellular fluid volume in hyponatremia. Am J Med1987; 83:905–988.
Hoorn EJ, Halperin ML, Zietse R. Diagnostic approach to a patient with hyponatraemia: traditional versus physiology-based options. QJM2005; 98:529–540.
Berl T. Impact of solute intake on urine flow and water excretion. J Am Soc Nephrol2008; 19:1076–1078.
Carrilho F, Bosch J, Arroyo V, Mas A, Viver J, Rodes J. Renal failure associated with demeclocycline in cirrhosis. Ann Intern Med1977; 87:195–197.
Lehrich RW, Greenberg A. When is it appropriate to use vasopressin receptor antagonists?J Am Soc Nephrol2008; 19:1054–1058.
Decaux G, Soupart A, Vassart G. Non-peptide arginine-vasopressin antagonists: the vaptans. Lancet2008; 371:1624–1632.
Schrier RW, Gross P, Gheorghiade M, et al; SALT Investigators. Tolvaptan, a selective oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl J Med2006; 355:2099–2112.
Konstam MA, Gheorghiade M, Burnett JC, et al; Efficacy of Vasopressin Antagonism in Heart Failure Outcome Study With Tolvaptan (EVEREST) Investigators. Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA2007; 297:1319–1331.
Berl T, Quittnat-Pelletier F, Verbalis JG, et al; SALTWATER Investigators. Oral tolvaptan is safe and effective in chronic hyponatremia. J Am Soc Nephrol2010; 21:705–712.
Greenberg A, Lehrich RW. Treatment of chronic hyponatremia: now we know how, but do we know when or if?J Am Soc Nephrol2010; 21:552–555.
Greenberg A, Verbalis JG. Vasopressin receptor antagonists. Kidney Int2006; 69:2124–2130.
Rosner MH, Kirven J. Exercise-associated hyponatremia. Clin J Am Soc Nephrol2007; 2:151–161.
Halperin ML, Kamel KS, Sterns R. Hyponatremia in marathon runners. N Engl J Med2005; 353:427–428.
Hew-Butler T, Almond C, Ayus JC, et al; Exercise-Associated Hyponatremia (EAH) Consensus Panel. Consensus statement of the 1st International Exercise-Associated Hyponatremia Consensus Development Conference, Cape Town, South Africa 2005. Clin J Sport Med2005; 15:208–213.
References
Flear CTG, Gill GV, Burn J. Hyponatremia: mechanisms and management. Lancet1981; 2:26–31.
Freda BJ, Davidson MB, Hall PM. Evaluation of hyponatremia: a little physiology goes a long way. Cleve Clin J Med2004; 71:639–650.
Weisberg LS. Pseudohyponatremia: a reappraisal. Am J Med1989; 86:315–318.
Moritz L, Ayus JC. The pathophysiology and treatment of hyponatraemic encephalopathy: an update. Nephrol Dial Transplant2003; 18:2486–2491.
Widdess-Walsh P, Sabharwal V, Demirjian S, DeGeorgia M. Neurologic effects of hyponatremia and its treatment. Cleve Clin J Med2007; 74:377–383.
Melton JE, Patlak CS, Pettigrew KD, Cserr HF. Volume regulatory loss of Na, Cl, and K from rat brain during acute hyponatremia. Am J Physiol1987; 252:F661–F669.
Lauriat SM, Berl T. The hyponatremic patient: practical focus on therapy. J Am Soc Nephrol1997; 8:1599–1607.
Kokko JP. Symptomatic hyponatremia with hypoxia is a medical emergency. Kidney Int2006; 69:1291–1293.
Ellis SJ. Severe hyponatraemia: complications and treatment. QJM1995; 88:905–909.
Sterns RH, Cappuccio JD, Silver SM, Cohen EP. Neurologic sequelae after treatment of severe hyponatremia: a multicenter perspective. J Am Soc Nephrol1994; 4:1522–1530.
Perianayagam A, Sterns RH, Silver SM, et al. DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia. Clin J Am Soc Nephrol2008; 3:331–336.
Sterns RH, Hix JK. Overcorrection of hyponatremia is a medical emergency. Kidney Int2009; 76:587–589.
Nguyen MK, Kurtz I. Analysis of current formulas used for treatment of the dysnatremias. Clin Exp Nephrol2004; 8:12–16.
Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med2000; 342:1581–1589.
Mohmand HK, Issa D, Ahmad Z, Cappuccio JD, Kouides RW, Sterns RH. Hypertonic saline for hyponatremia: risk of inadvertent overcorrection. Clin J Am Soc Nephrol2007; 2:1110–1117.
Ellison DH, Berl T. Clinical practice. The syndrome of inappropriate antidiuresis. N Engl J Med2007; 356:2064–2072.
Renneboog B, Musch W, Vandemergel X, Manto MU, Decaux G. Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits. Am J Med2006; 119:71.e1–e8.
Kinsella S, Moran S, Sullivan MO, Molloy MG, Eustace JA. Hyponatremia independent of osteoporosis is associated with fracture occurrence. Clin J Am Soc Nephrol2010; 5:275–280.
Chung HM, Kluge R, Schrier RW, Anderson RJ. Clinical assessment of extracellular fluid volume in hyponatremia. Am J Med1987; 83:905–988.
Hoorn EJ, Halperin ML, Zietse R. Diagnostic approach to a patient with hyponatraemia: traditional versus physiology-based options. QJM2005; 98:529–540.
Berl T. Impact of solute intake on urine flow and water excretion. J Am Soc Nephrol2008; 19:1076–1078.
Carrilho F, Bosch J, Arroyo V, Mas A, Viver J, Rodes J. Renal failure associated with demeclocycline in cirrhosis. Ann Intern Med1977; 87:195–197.
Lehrich RW, Greenberg A. When is it appropriate to use vasopressin receptor antagonists?J Am Soc Nephrol2008; 19:1054–1058.
Decaux G, Soupart A, Vassart G. Non-peptide arginine-vasopressin antagonists: the vaptans. Lancet2008; 371:1624–1632.
Schrier RW, Gross P, Gheorghiade M, et al; SALT Investigators. Tolvaptan, a selective oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl J Med2006; 355:2099–2112.
Konstam MA, Gheorghiade M, Burnett JC, et al; Efficacy of Vasopressin Antagonism in Heart Failure Outcome Study With Tolvaptan (EVEREST) Investigators. Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA2007; 297:1319–1331.
Berl T, Quittnat-Pelletier F, Verbalis JG, et al; SALTWATER Investigators. Oral tolvaptan is safe and effective in chronic hyponatremia. J Am Soc Nephrol2010; 21:705–712.
Greenberg A, Lehrich RW. Treatment of chronic hyponatremia: now we know how, but do we know when or if?J Am Soc Nephrol2010; 21:552–555.
Greenberg A, Verbalis JG. Vasopressin receptor antagonists. Kidney Int2006; 69:2124–2130.
Rosner MH, Kirven J. Exercise-associated hyponatremia. Clin J Am Soc Nephrol2007; 2:151–161.
Halperin ML, Kamel KS, Sterns R. Hyponatremia in marathon runners. N Engl J Med2005; 353:427–428.
Hew-Butler T, Almond C, Ayus JC, et al; Exercise-Associated Hyponatremia (EAH) Consensus Panel. Consensus statement of the 1st International Exercise-Associated Hyponatremia Consensus Development Conference, Cape Town, South Africa 2005. Clin J Sport Med2005; 15:208–213.
Some hyponatremic patients present with acute, life-threatening cerebral edema due to severe hyponatremia. In others, the hyponatremia may be chronic and less severe, causing relatively few symptoms, but representing an important, independent marker of poor prognosis due to an underlying disease (eg, heart failure).
Even patients with chronic, less severe hyponatremia may have subtle symptoms of neurocognitive dysfunction and a higher risk of bone fractures.
Overly rapid correction of chronic hyponatremia or undercorrection of acute symptomatic hyponatremia can lead to serious neurologic injury.
Treatment strategies vary depending on the extracellular fluid volume status and the cause of hyponatremia.
Vasopressin antagonists (“vaptans”), a new class of aquaretic agents, specifically target the mechanism driving hyponatremia in some patients.
No. There is no scientific basis for recommending vitamin-mineral supplements to the healthy population.
(This commentary deals only with healthy people in the general US population. There are well-established guidelines for the use of supplements in pregnant and lactating women, infants, and individuals with a wide variety of health conditions.)
TAKE A PILL, OR EAT A HEALTHIER DIET?
The Dietary Guidelines for Americans1 reported that the US population consumes insufficient amounts of green leafy vegetables, fresh fruits, whole grains, and fiber and excessive amounts of refined carbohydrates, saturated fat, and sodium. This may result in inadequate intake of some nutrients. (The term “inadequate” intake is being used to differentiate this situation from “deficiency,” which is rare in the general population.)
So the real question is, Should we pop a vitamin pill every day and forget about it, or try to eat a healthier diet?
To answer that question, consider this: no supplement trial has ever been able to reproduce the health benefits of eating adequate amounts of fresh fruits and vegetables.
One reason is that natural foods contain far more compounds than the few we know about and can put in a supplement pill. For example, vegetables contain hundreds of antioxidant compounds, many perhaps acting synergistically, while so far we have been able to identify and isolate only a handful.
Second, nutrients have different health effects depending on the host’s conditions. A calcium supplement will not increase bone mineral density unless accompanied by regular, weight-bearing exercise that stimulates bone accretion.
This is why most supplement trials have shown disappointing results. A recent National Institutes of Health state-of-the-science conference on multivitamin-mineral supplements2 concluded that there is no consistent evidence that single-vitamin or multivitamin supplements help in preventing a wide range of diseases studied.
‘AT LEAST IT WON’T HURT’ MAY NOT BE TRUE
In spite of the lack of evidence, many will go on taking supplements, with the argument that “at least it won’t hurt.” They should be reminded that several supplement trials had to be stopped prematurely due to unexpected adverse effects.
In the Selenium and Vitamin E Cancer Prevention Trial (SELECT),3 which evaluated supplementation to prevent prostate cancer, the group receiving vitamin E had more cases of prostate cancer than controls, and the group taking selenium had more diabetes cases. While these differences were not statistically significant, they were of enough concern to stop the trial.
A meta-analysis of vitamin E trials4 showed a slight increase in the rate of all-cause mortality in those receiving the active supplement.
The bottom line: the evidence that supplements “won’t hurt” is even more limited than the evidence for their efficacy, because trials are usually not designed to address safety outcomes.
TELL YOUR PATIENTS THE THINGS THEY DO NOT WANT TO HEAR
Unfortunately, this means you have to tell your patients all the things they do not want to hear: cut the ice cream, eat more broccoli, exercise regularly. But because of their position of authority and credibility, physicians can play a crucial role in helping the US population improve its dietary and lifestyle habits.
The key is to introduce and support minor but sustained changes in the diet and physical activity. For example, we have shown that simply reducing consumption of caloric beverages (soft drinks) can result in significant weight loss in overweight adults, without any other dietary intervention.5
The other key is of course to modify the obesogenic environment we live in. Only by creating conditions that facilitate healthy eating and regular activity will we have a significant impact on public health.
References
United States Department of Agriculture Center for Nutrition Policy and Promotion. Report of the Dietary Guidelines Advisory Committee on the Dietary Guidelines for Americans, 2010. www.cnpp.usda.gov/DGAs2010-DGACReport.htm. Accessed 8/25/2010.
Huang HY, Caballero B, Chang S, et al. The efficacy and safety of multivitamin and mineral supplement use to prevent cancer and chronic disease in adults: a systematic review for a National Institutes of Health state-of-the-science conference. Ann Intern Med2006; 145:372–385.
Lippman SM, Klein EA, Goodman PJ, et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA2009; 301:39–51.
Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med2005; 142:37–46.
Chen L, Appel LJ, Loria C, et al. Reduction in consumption of sugar-sweetened beverages is associated with weight loss: the PREMIER trial. Am J Clin Nutr2009; 89:1299–1306.
Benjamin Caballero, MD, PhD Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
Address: Benjamin Caballero, MD, PhD, Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Rm. 2041, Baltimore, MD 21205; e-mail [email protected]
Benjamin Caballero, MD, PhD Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
Address: Benjamin Caballero, MD, PhD, Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Rm. 2041, Baltimore, MD 21205; e-mail [email protected]
Author and Disclosure Information
Benjamin Caballero, MD, PhD Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
Address: Benjamin Caballero, MD, PhD, Professor of Pediatrics, Nutrition, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Rm. 2041, Baltimore, MD 21205; e-mail [email protected]
No. There is no scientific basis for recommending vitamin-mineral supplements to the healthy population.
(This commentary deals only with healthy people in the general US population. There are well-established guidelines for the use of supplements in pregnant and lactating women, infants, and individuals with a wide variety of health conditions.)
TAKE A PILL, OR EAT A HEALTHIER DIET?
The Dietary Guidelines for Americans1 reported that the US population consumes insufficient amounts of green leafy vegetables, fresh fruits, whole grains, and fiber and excessive amounts of refined carbohydrates, saturated fat, and sodium. This may result in inadequate intake of some nutrients. (The term “inadequate” intake is being used to differentiate this situation from “deficiency,” which is rare in the general population.)
So the real question is, Should we pop a vitamin pill every day and forget about it, or try to eat a healthier diet?
To answer that question, consider this: no supplement trial has ever been able to reproduce the health benefits of eating adequate amounts of fresh fruits and vegetables.
One reason is that natural foods contain far more compounds than the few we know about and can put in a supplement pill. For example, vegetables contain hundreds of antioxidant compounds, many perhaps acting synergistically, while so far we have been able to identify and isolate only a handful.
Second, nutrients have different health effects depending on the host’s conditions. A calcium supplement will not increase bone mineral density unless accompanied by regular, weight-bearing exercise that stimulates bone accretion.
This is why most supplement trials have shown disappointing results. A recent National Institutes of Health state-of-the-science conference on multivitamin-mineral supplements2 concluded that there is no consistent evidence that single-vitamin or multivitamin supplements help in preventing a wide range of diseases studied.
‘AT LEAST IT WON’T HURT’ MAY NOT BE TRUE
In spite of the lack of evidence, many will go on taking supplements, with the argument that “at least it won’t hurt.” They should be reminded that several supplement trials had to be stopped prematurely due to unexpected adverse effects.
In the Selenium and Vitamin E Cancer Prevention Trial (SELECT),3 which evaluated supplementation to prevent prostate cancer, the group receiving vitamin E had more cases of prostate cancer than controls, and the group taking selenium had more diabetes cases. While these differences were not statistically significant, they were of enough concern to stop the trial.
A meta-analysis of vitamin E trials4 showed a slight increase in the rate of all-cause mortality in those receiving the active supplement.
The bottom line: the evidence that supplements “won’t hurt” is even more limited than the evidence for their efficacy, because trials are usually not designed to address safety outcomes.
TELL YOUR PATIENTS THE THINGS THEY DO NOT WANT TO HEAR
Unfortunately, this means you have to tell your patients all the things they do not want to hear: cut the ice cream, eat more broccoli, exercise regularly. But because of their position of authority and credibility, physicians can play a crucial role in helping the US population improve its dietary and lifestyle habits.
The key is to introduce and support minor but sustained changes in the diet and physical activity. For example, we have shown that simply reducing consumption of caloric beverages (soft drinks) can result in significant weight loss in overweight adults, without any other dietary intervention.5
The other key is of course to modify the obesogenic environment we live in. Only by creating conditions that facilitate healthy eating and regular activity will we have a significant impact on public health.
No. There is no scientific basis for recommending vitamin-mineral supplements to the healthy population.
(This commentary deals only with healthy people in the general US population. There are well-established guidelines for the use of supplements in pregnant and lactating women, infants, and individuals with a wide variety of health conditions.)
TAKE A PILL, OR EAT A HEALTHIER DIET?
The Dietary Guidelines for Americans1 reported that the US population consumes insufficient amounts of green leafy vegetables, fresh fruits, whole grains, and fiber and excessive amounts of refined carbohydrates, saturated fat, and sodium. This may result in inadequate intake of some nutrients. (The term “inadequate” intake is being used to differentiate this situation from “deficiency,” which is rare in the general population.)
So the real question is, Should we pop a vitamin pill every day and forget about it, or try to eat a healthier diet?
To answer that question, consider this: no supplement trial has ever been able to reproduce the health benefits of eating adequate amounts of fresh fruits and vegetables.
One reason is that natural foods contain far more compounds than the few we know about and can put in a supplement pill. For example, vegetables contain hundreds of antioxidant compounds, many perhaps acting synergistically, while so far we have been able to identify and isolate only a handful.
Second, nutrients have different health effects depending on the host’s conditions. A calcium supplement will not increase bone mineral density unless accompanied by regular, weight-bearing exercise that stimulates bone accretion.
This is why most supplement trials have shown disappointing results. A recent National Institutes of Health state-of-the-science conference on multivitamin-mineral supplements2 concluded that there is no consistent evidence that single-vitamin or multivitamin supplements help in preventing a wide range of diseases studied.
‘AT LEAST IT WON’T HURT’ MAY NOT BE TRUE
In spite of the lack of evidence, many will go on taking supplements, with the argument that “at least it won’t hurt.” They should be reminded that several supplement trials had to be stopped prematurely due to unexpected adverse effects.
In the Selenium and Vitamin E Cancer Prevention Trial (SELECT),3 which evaluated supplementation to prevent prostate cancer, the group receiving vitamin E had more cases of prostate cancer than controls, and the group taking selenium had more diabetes cases. While these differences were not statistically significant, they were of enough concern to stop the trial.
A meta-analysis of vitamin E trials4 showed a slight increase in the rate of all-cause mortality in those receiving the active supplement.
The bottom line: the evidence that supplements “won’t hurt” is even more limited than the evidence for their efficacy, because trials are usually not designed to address safety outcomes.
TELL YOUR PATIENTS THE THINGS THEY DO NOT WANT TO HEAR
Unfortunately, this means you have to tell your patients all the things they do not want to hear: cut the ice cream, eat more broccoli, exercise regularly. But because of their position of authority and credibility, physicians can play a crucial role in helping the US population improve its dietary and lifestyle habits.
The key is to introduce and support minor but sustained changes in the diet and physical activity. For example, we have shown that simply reducing consumption of caloric beverages (soft drinks) can result in significant weight loss in overweight adults, without any other dietary intervention.5
The other key is of course to modify the obesogenic environment we live in. Only by creating conditions that facilitate healthy eating and regular activity will we have a significant impact on public health.
References
United States Department of Agriculture Center for Nutrition Policy and Promotion. Report of the Dietary Guidelines Advisory Committee on the Dietary Guidelines for Americans, 2010. www.cnpp.usda.gov/DGAs2010-DGACReport.htm. Accessed 8/25/2010.
Huang HY, Caballero B, Chang S, et al. The efficacy and safety of multivitamin and mineral supplement use to prevent cancer and chronic disease in adults: a systematic review for a National Institutes of Health state-of-the-science conference. Ann Intern Med2006; 145:372–385.
Lippman SM, Klein EA, Goodman PJ, et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA2009; 301:39–51.
Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med2005; 142:37–46.
Chen L, Appel LJ, Loria C, et al. Reduction in consumption of sugar-sweetened beverages is associated with weight loss: the PREMIER trial. Am J Clin Nutr2009; 89:1299–1306.
References
United States Department of Agriculture Center for Nutrition Policy and Promotion. Report of the Dietary Guidelines Advisory Committee on the Dietary Guidelines for Americans, 2010. www.cnpp.usda.gov/DGAs2010-DGACReport.htm. Accessed 8/25/2010.
Huang HY, Caballero B, Chang S, et al. The efficacy and safety of multivitamin and mineral supplement use to prevent cancer and chronic disease in adults: a systematic review for a National Institutes of Health state-of-the-science conference. Ann Intern Med2006; 145:372–385.
Lippman SM, Klein EA, Goodman PJ, et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA2009; 301:39–51.
Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med2005; 142:37–46.
Chen L, Appel LJ, Loria C, et al. Reduction in consumption of sugar-sweetened beverages is associated with weight loss: the PREMIER trial. Am J Clin Nutr2009; 89:1299–1306.
The process may be relatively easy for the few diseases in which an etiologic agent fulfills Koch’s postulates. Otherwise, potential targets for therapeutic intervention are selected via several approaches.
Observational studies may suggest risk factors, and a number of molecular techniques may be used to identify specific cell types, up-regulated genes, or overexpressed potential critical mediators within diseased tissues. The latter approach was used, in part, in the successful development of therapies for rheumatoid arthritis that block tumor necrosis factor (TNF) and interleukin 6 (IL-6).
Once a potential target is found, molecular biologists using current tools of drug development—gene chip analysis, molecular modeling, proteomic scanning, and hybridoma-based synthesis—can produce a small molecule or biologic product to block the effect or expression of nearly any molecule or pathway (although a successful therapy cannot always be developed easily).
But sometimes, potential markers of disease pathogenesis are actually embers of the pathologic process rather than flames driving the disease.
For example, consider tuberculosis. Granulomas are typical of Mycobacterium tuberculosis infection. Suppose we didn’t know about mycobacteria but assumed instead it was the granuloma per se that was the actual agent of disease. It might then be reasonable to try to treat tuberculosis with the anti-TNF agents, since they can prevent the development of mature granulomas. But that approach would lead to uncontrolled M tuberculosis infection, not resolution of clinical tuberculosis.
Although not entirely analogous to an infectious disease, Alzheimer disease is another case in point. On page 689 in this issue of the Journal,Dr. David S. Geldmacher discusses how the amyloid plaques of Alzheimer disease may be the footprint but not the actual cause of the disorder. This hypothesis would explain the failure of attempts at treating Alzheimer disease by attacking this presumed pathogenic target and, if true, would require starting anew to find the right target.
The process may be relatively easy for the few diseases in which an etiologic agent fulfills Koch’s postulates. Otherwise, potential targets for therapeutic intervention are selected via several approaches.
Observational studies may suggest risk factors, and a number of molecular techniques may be used to identify specific cell types, up-regulated genes, or overexpressed potential critical mediators within diseased tissues. The latter approach was used, in part, in the successful development of therapies for rheumatoid arthritis that block tumor necrosis factor (TNF) and interleukin 6 (IL-6).
Once a potential target is found, molecular biologists using current tools of drug development—gene chip analysis, molecular modeling, proteomic scanning, and hybridoma-based synthesis—can produce a small molecule or biologic product to block the effect or expression of nearly any molecule or pathway (although a successful therapy cannot always be developed easily).
But sometimes, potential markers of disease pathogenesis are actually embers of the pathologic process rather than flames driving the disease.
For example, consider tuberculosis. Granulomas are typical of Mycobacterium tuberculosis infection. Suppose we didn’t know about mycobacteria but assumed instead it was the granuloma per se that was the actual agent of disease. It might then be reasonable to try to treat tuberculosis with the anti-TNF agents, since they can prevent the development of mature granulomas. But that approach would lead to uncontrolled M tuberculosis infection, not resolution of clinical tuberculosis.
Although not entirely analogous to an infectious disease, Alzheimer disease is another case in point. On page 689 in this issue of the Journal,Dr. David S. Geldmacher discusses how the amyloid plaques of Alzheimer disease may be the footprint but not the actual cause of the disorder. This hypothesis would explain the failure of attempts at treating Alzheimer disease by attacking this presumed pathogenic target and, if true, would require starting anew to find the right target.
Picking a therapeutic target is not always easy.
The process may be relatively easy for the few diseases in which an etiologic agent fulfills Koch’s postulates. Otherwise, potential targets for therapeutic intervention are selected via several approaches.
Observational studies may suggest risk factors, and a number of molecular techniques may be used to identify specific cell types, up-regulated genes, or overexpressed potential critical mediators within diseased tissues. The latter approach was used, in part, in the successful development of therapies for rheumatoid arthritis that block tumor necrosis factor (TNF) and interleukin 6 (IL-6).
Once a potential target is found, molecular biologists using current tools of drug development—gene chip analysis, molecular modeling, proteomic scanning, and hybridoma-based synthesis—can produce a small molecule or biologic product to block the effect or expression of nearly any molecule or pathway (although a successful therapy cannot always be developed easily).
But sometimes, potential markers of disease pathogenesis are actually embers of the pathologic process rather than flames driving the disease.
For example, consider tuberculosis. Granulomas are typical of Mycobacterium tuberculosis infection. Suppose we didn’t know about mycobacteria but assumed instead it was the granuloma per se that was the actual agent of disease. It might then be reasonable to try to treat tuberculosis with the anti-TNF agents, since they can prevent the development of mature granulomas. But that approach would lead to uncontrolled M tuberculosis infection, not resolution of clinical tuberculosis.
Although not entirely analogous to an infectious disease, Alzheimer disease is another case in point. On page 689 in this issue of the Journal,Dr. David S. Geldmacher discusses how the amyloid plaques of Alzheimer disease may be the footprint but not the actual cause of the disorder. This hypothesis would explain the failure of attempts at treating Alzheimer disease by attacking this presumed pathogenic target and, if true, would require starting anew to find the right target.