The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Published Online:

Abstract

Cardiovascular disease (CVD) affects one in five Americans and is the leading cause of death in the United States, claiming more lives than the next five leading causes of death combined. Psychiatric disorders are also quite common: anxiety disorders affect 20 million people annually, and depression affects 17 million people. CVD and psychiatric illness often coexist. Anxiety and depression are more prevalent in patients with CVD than in the general population. Depression has long been linked to poor medical compliance, to other risk factors for CVD such as smoking and obesity, and to greater functional impairment. Depression also independently predicts the development of CVD in the general population, as well as future cardiac events and mortality in patients with CVD. Anxiety or anxiety disorders independently predict sudden cardiac death in the general population as well as future cardiac events in patients with CVD. The treatment of psychiatric disorders in patients with CVD can be challenging because of the cardiovascular side effects of many psychotropic medications as well as the potential of multiple drug-drug interactions. Moreover, many medications for CVD have psychiatric side effects. This review covers a wide variety of issues related to CVD and psychiatry and provides a guide for psychiatrists treating patients with comorbid CVD and psychiatric symptoms.

Worry affects the circulation, the heart, the glands, the whole nervous system. I have never known a man who died from overwork, but many who died from doubt.

—Charles Horace Mayo (1865–1939)

The heart has its reasons which reason does not know.

—Blaise Pascal (1623–1662)

Cardiovascular disease (CVD) is the leading cause of death in the United States, claiming more lives than the next five leading causes of death combined—among them cancer, accidents, and diabetes. One in five men and women in the United States have some form of CVD, and in 2001, one of every 2.6 deaths was attributable to CVD. For 2004, the direct and indirect cost of CVD was estimated at $368.4 billion (1). Inasmuch as anxiety disorders affect 20 million Americans annually and depression 17 million (2, 3), it is not surprising that there is extensive comorbidity of psychiatric disorders and cardiovascular disease. Yet there appears to be more to the relationship of mental disorders with heart disease than a coincidental occurrence. Mood and anxiety problems may be risk factors for heart disease, and the occurrence of heart disease both increases the risk and complicates the treatment of psychiatric illness.

Evidence for the association of depression with cardiovascular disease first emerged in studies of institutionalized psychiatric patients more than 70 years ago (4, 5). Anecdotal reports of anger- and anxiety-induced cardiac events also provided a basis for speculation about mind-body relationships in heart disease, an issue addressed further in Cannon’s work on voodoo death (6). In recent years, there has been a dramatic increase in research about the relationship between psychiatric symptoms and disorders and cardiovascular disease. Such research has been spurred by several factors:

1.

Improved epidemiological and diagnostic methods

2.

New knowledge of the effects of the autonomic nervous system and other physiological consequences of stress and anxiety

3.

Increasing integration of biological and psychological conceptualizations of mental disorders

4.

The recognition of the cardiovascular effects of psychotropic drugs

This article focuses primarily on depression and anxiety, which have been the most studied psychiatric conditions in relation to cardiovascular disease. We will also discuss research on personality traits, acute stress, and other psychiatric symptoms or conditions that have been studied in relation to coronary heart disease and heart failure.

Epidemiology of psychiatric problems in heart disease patients

Depression

Cardiac disease has an enormous impact on an individual’s overall health and daily functioning. Cardiac events often result in disability and a change in social role function and affect the individual’s perception of his or her mortality. Hence it is not surprising that depression appears to be the most common psychiatric disorder in patients with coronary artery disease (CAD) (4). Estimates of the prevalence of depression in post–myocardial infarction (MI) patients in studies in the 1960s and 1970s ranged from 10% to 87%. The breadth of this range is probably attributable to wide variations in diagnostic criteria, in the time windows after MI in which depression was measured, and in the studies’ inclusion and exclusion criteria (5). More recent studies consistently indicate a prevalence of post-MI depression in the range of 16%–27% (711). The Montreal Heart Institute study found the 1-year cumulative incidence of depression in 218 post-MI patients to be 31% (12), a result that was recently reproduced in an expanded sample (13). A study of 200 patients who had suffered a first MI found a 1-year cumulative incidence of depression of 25% (14).

In inpatients as well as outpatients with CAD, studies have placed the point prevalence of major depressive disorder within the range of 15%–24% (1519). In the first few weeks or months following coronary artery bypass graft (CABG) surgery, the point prevalence of depression is in the range of 20%–30% (2022). In patients with congestive heart failure (CHF), three studies have found the prevalence of depression to be approximately 20% (2325), and one study found a 35% point prevalence of depression in 374 CHF patients admitted to an inpatient cardiology service (24).

Anxiety

The epidemiology of anxiety disorders in CAD is not as well studied as that of depression, even though symptoms of anxiety are relatively common in cardiac patients. The incidence of anxiety symptoms in patients with acute coronary disease in cardiac care units is approximately 50% (26, 27). Anxiety symptoms have been reported to be elevated in 5%–10% of patients with chronic heart disease (28). A recent study of 100 stable outpatients with coronary heart disease (CHD) using structured interviews for DSM-IV diagnosis demonstrated that multiple psychiatric diagnoses were common. Generalized anxiety disorder and posttraumatic stress disorder (PTSD) were identified in more than 20% of patients. Alcohol abuse, recurrent major depression, past episodes of depression, and current dysthymia were also prevalent, but single-episode current major depression was rare (29). Of 61 post-MI patients referred to a cardiac rehabilitation program, 4.9% were diagnosed with social phobia and 3.3% with agoraphobia without panic attacks (30). Lane and colleagues, in a study of 288 patients hospitalized for MI, found a 26% incidence of high levels of state anxiety (STAI≥40) (31). In a study of CABG patients, 55% had high levels of state anxiety preoperatively, and 32% had clinically significant levels of anxiety 3 months after surgery (32).

There are few data on the prevalence of anxiety disorders in patients with CHF (33). In a review of data from the Studies of Left Ventricular Dysfunction (SOLVD) trials, women were found to have significantly worse anxiety than women in several comparison groups, including normal subjects, geriatric subjects, hypertension patients, and cancer patients (34). High levels of anxiety symptoms were also reported in subjects in a survey of ambulatory outpatients with dilated cardiomyopathy (35).

Patients with implanted cardioverter-defibrillators (ICDs) are a special subgroup with a high risk of anxiety problems. ICDs, approved for use by the Food and Drug Administration in 1985, automatically deliver electrical shocks to the heart to interrupt abnormal rhythms; the device improves survival in patients at risk of developing lethal arrhythmias (36). The shock from the ICD is strong and unpleasant, a sensation sometimes described as a “kick in the chest.” Discharges may occur without any antecedent subjective symptoms, and the presence of the device is itself a stressor for many patients, especially those who have already experienced a discharge. After experiencing shocks, symptoms of PTSD, such as avoidance, hypervigilance, and reexperiencing, are common. This is especially true if patients experience multiple sequential shocks while conscious, which they may endure with a sense of helplessness. Some patients with ICDs meet criteria for full PTSD (37).

In an early study of psychiatric morbidity in patients with ICDs, Morris and colleagues found a 50% prevalence of psychiatric disorders (adjustment disorders, major depression, and panic disorder) in 20 patients examined 3 to 21 months after the ICD implant (38). Chevalier and colleagues reported a 20% prevalence of anxiety disorders in 30 patients (39). Although one study found that no new anxiety disorders developed in ICD patients by 9–18 months after implantation (40), Bourke and associates found that over 6 years, 6 of 35 patients demonstrated “florid” psychological problems, with anxiety symptoms, including two cases of generalized anxiety disorder, two cases of agoraphobia with panic, and two cases of major depression (41). Similarly, in a study of 72 ICD patients evaluated 1 to 6 years after implantation, Godemann et al. found that 14 patients (19.4%) had agoraphobia and/or panic disorder, 11 of whom developed the anxiety disorder only after ICD placement (42).

Recent large-scale outcome studies of ICD therapy for ventricular arrhythmias confirm that discharges that are repetitive or frequent or occur soon after implantation are associated with reduced mental well-being, reduced physical function, and increased anxiety (43, 44). One recent trial of defibrillator-delivered pacing and shock therapy for atrial fibrillation found no significant difference in perceived quality of life between those who had and those who had not received shocks (45). Similarly, Duru and colleagues found no differences in symptoms of anxiety and depression between pacemaker patients and ICD patients, regardless of whether or not they had received shocks (46).

Impact of psychological factors on cardiovascular disease

Depression and known cardiac risk factors

Depression is linked to a large number of major risk factors for CAD or cardiac-related mortality, including cigarette smoking, diabetes, and obesity. The statistical overlap between depression and having one or more of these risk factors has long been known. Prospective studies indicate that depression is an independent risk factor in the development of these behaviors or conditions.

Cigarette smoking is clearly associated with an increased risk of CVD. Current smokers have almost three times the rate of acute MI when compared with those who have never smoked (47). Depression is independently linked to daily smoking and nicotine dependence (48). Researchers have found that people with a history of major depression have a threefold elevation in risk of becoming a smoker (49). Depressed smokers are less likely to quit successfully and are more likely to have withdrawal symptoms during attempts to quit (5052). Smokers with a history of depression have been shown to have an exaggerated belief in the positive effects of smoking (53). They also have less confidence in their ability to refrain from smoking (54).

Diabetes is associated with a three- to fourfold increase in risk of developing CVD and cardiovascular mortality (55). Depression is a risk factor for developing diabetes even after other cardiac risk factors are controlled for. An 8-year prospective study of 2,764 male employees found that subjects who had a moderate or severe level of depressive symptoms had 2.3 times the risk of developing type 2 diabetes compared with those who had no depressive symptoms (56). Two large prospective studies have recently been published that further support this finding. The Atherosclerosis Risk in Communities (ARIC) study included 11,615 initially nondiabetic adults 48–67 years of age who were followed for 6 years for the development of type 2 diabetes. After adjustment for age, race, sex, and education, the risk of individuals in the highest quartile of depressive symptoms was 1.6 times that of those in the lowest quartile. Even after adjustment for lifestyle factors such as smoking and metabolic factors such as weight and blood pressure, the risk was still 1.4 times that of the nondepressed subjects (57). A prospective study of 72,178 female nurses 45–72 years of age who were followed for 4 years found that the age-adjusted risk of developing type 2 diabetes among women with depressive symptoms was 1.55. After adjustment for body mass index (BMI) the risk was 1.36, a finding consistent with the ARIC study results (58).

Depression is also linked to the development of obesity, another CVD risk factor. One prospective study of patients 6–17 years of age found that after controlling for other risk factors, including age, cigarette use, and socioeconomic class, there was a significant correlation between childhood depression and BMI 10–15 years later. Those who had major depression had a BMI±SD of 26.1±5.2, while those without depression had a BMI 24.2±4.1 (59). A prospective cohort study of 9,374 adolescents in grades 7 through 12 assessed via in-home interviews and self-reported height and weight found at 1-year follow-up that depressed mood at baseline independently raised the risk of obesity twofold, even after potential confounders were controlled for. Baseline obesity did not predict follow-up depression even though depression did predict obesity (60).

Depression as an independent coronary disease risk factor

Even after the effects of depression on health behaviors, smoking, diabetes, and weight are taken into account, there is ample evidence that depressive symptoms and a history of depression are strong independent risk factors for the development of cardiovascular disease, of acute coronary events, and of mortality from cardiac illness. In general, studies show that the relative risk of incident cardiac disease in healthy individuals with depression or symptoms of depression is about 1.5 to 2, depending on which cardiac endpoint is used. The risk tends to be graduated, increasing with the level of depression (5, 61, 62). A full clinical diagnosis of depression yields a higher risk of developing CHD. The Mini-Finland Health Survey, which followed subjects for an average of 6.6 years, demonstrated a relative risk of 3.36 for the development of CHD in those with major depression (63). A follow-up of the Baltimore Epidemiologic Catchment Area (ECA) study that looked at 1,551 subjects without heart disease over a 13-year period found that those with a history of a major depressive episode had an odds ratio for having a nonfatal MI of 4.54 when compared with those without a history of major depression or dysphoria (64).

The impact of depression on the prognosis of patients with preexisting cardiac disease has been extensively studied. In the relatively few studies of patients with verifiable, established CAD (e.g., diagnosed by angiography) but without any recent cardiac event, depression has been found to be predictive of future cardiac mortality and morbidity (6567). Depression has also been found to be a risk factor for mortality after CABG, in follow-up periods ranging from 2 to 12 years (20, 68, 69).

In post-MI patients, probably the most widely cited study is that of Frasure-Smith and colleagues (10, 70). In 222 patients, they demonstrated that the risk of death in the first 6 months after MI for patients with major depression was five times that of the nondepressed patients (odds ratio, 4.29) (9). While patients with depressive symptoms (Beck Depression Inventory score ≥ 10) but without major depressive disorder did not have an elevated risk at 6 months, by 18 months they had an odds ratio of 7.82 compared with those with a Beck Depression Inventory score < 10. For patients with Beck Depression Inventory ≥ 10 and more than nine premature ventricular contractions per hour, the odds ratio for mortality was 29.17 compared with other patients (70). Lesperance and colleagues followed 896 post-MI patients for 5 years, evaluating depression during admission and again 1 year after MI, and found a significant dose-response relationship between initial in-hospital depressive symptoms and cardiac mortality. Depressive symptoms at 1 year were not independently correlated with cardiac mortality. Patients with mild to moderate depression symptoms at baseline who had reduced depression symptoms at 1-year follow-up went on to demonstrate lower cardiac mortality, while patients with high depression symptoms at baseline did not. This raises the question of whether there are distinct subtypes of depression with different prognostic implications (71).

Not all studies have had such clearly positive results. Irvine and colleagues found that the apparent twofold increase in risk of mortality in a 2-year follow-up of post-MI patients was reduced by 30% and no longer statistically significant after controlling for dyspnea and fatigue (72). Strik and colleagues looked at 318 men who completed questionnaires on anxiety, depression, and hostility after MI and found that while depression seemed to be associated with a higher risk of cardiac events over an average of 3.4 years of follow-up, anxiety symptoms alone were the independent predictor of cardiac events (73). The seemingly positive results of two other studies, both with low statistical power, also did not survive risk factor adjustment (10, 74).

In patients with CHF, studies have shown that depression is associated with a higher risk of mortality. Murberg and colleagues evaluated 119 stable outpatients with heart failure and found that depressed mood doubled the risk of mortality (25% vs. 11.3%), with a significant difference remaining even after other factors were controlled for (75). In 374 inpatients with CHF, a Beck Depression Inventory ≥ 10 combined with a diagnosis of major depression on interview significantly increased mortality at 1-year follow-up (24). After other risk factors were controlled for, only increased readmission rates at 1 year persisted as a significant difference (24). A 5-year follow-up assessment of 396 inpatients with heart failure due to dilated cardiomyopathy found that the patients with clinical depression had a threefold greater risk of mortality (25). Major depression was associated with increased mortality at 1 year in 374 inpatients with CHF (76). Low emotional support was associated with worse 1-year cardiac outcome in a sample of 168 female elderly patients admitted with heart failure, although no such association was noted in the 124 male patients (77). Another study, however, failed to find an effect of depression on survival in hospitalized CHF patients after adjustment for illness severity (78).

Among patients who have depression after a cardiac event, those with a history of prior depression appear to have the greatest risk of mortality. Lesperance et al. (12) studied patients with depression after MI and found that at 1 year, 40% of those with a prior history of depression had died, compared with only 10% of those who experienced their first episode of depression after the cardiac event.

Anxiety

Three community-based studies have shown a significant relationship between anxiety disorders and sudden cardiac death. One study of 1,457 male subjects followed over 6 years found the risk of cardiac-related death elevated almost fourfold in those with phobic anxiety (79). In a study of 33,999 middle-aged male health professionals followed over 2 years, a dose-dependent relationship was observed between degree of phobic anxiety and risk of cardiac-related death. Highly anxious subjects had a sixfold elevation in risk of sudden cardiac death (80). In a study of 2,271 men followed over 32 years, anxiety was associated with a 1.9-fold increase in risk of fatal cardiac disease and a 4.7-fold increase in risk of sudden cardiac death, in a dose-dependent fashion (81). All three studies showed no link between anxiety and MI. Thus, anxiety-related cardiac mortality risk seems to be confined to sudden cardiac death, at least in males. This suggests that the predominant mechanism by which anxiety disorders increase risk in nonclinical samples is by inducing ventricular arrhythmias and not by increasing the risk of atherosclerosis.

Patients with preexisting cardiac disease, however, are in a different risk category than the general population. The presence of anxiety at a baseline cardiac event significantly predicts future MIs and cardiac events, not just sudden cardiac death. Frasure-Smith and colleagues (82) followed 222 post-MI patients for 12 months and found that elevated levels of self-reported state and trait anxiety at baseline were significantly and independently linked both to the occurrence of another MI and the development of any new cardiac event. Moser and Dracup (26) looked at anxiety in 86 new-MI patients within 48 hours of admission to the hospital, using a brief self-rating instrument. In-hospital complications occurred in 19.6% of those with high levels of anxiety symptoms, compared with 6% in those with low levels. Even after other risk factors were controlled for, patients with high anxiety still had almost a fivefold elevation in risk of cardiac complications or death compared with those with little or no anxiety. Denollet and Brutsaert studied 87 middle-aged post-MI patients with a left ventricular ejection fraction of 50% or less who were psychologically evaluated at baseline and then followed up an average of 8 years later. A high level of anxiety symptoms was not an independent risk factor for recurrent events after adjustment for other risk factors, but the combination of high anxiety and a high level of social inhibition was associated with an independent relative risk of 4.7 for having a new cardiac event (83).

Acute and chronic stress

Acutely stressful events dramatically increase cardiac morbidity and mortality. In a prospective study of 95,647 Finnish men and women, the risk of all-cause mortality in the first week following spousal death was two times normal. The risk of any ischemic heart disease event in that period was elevated 2.3-fold in men and 3.5-fold in women, an effect independent of age; the risk diminished dramatically by the end of the first month (84). The Northridge, California, earthquake of 1994 was followed by a 35% rise in hospitalizations for acute MI (85) and an almost fivefold increase in incidence of sudden cardiac death (86). A study on cardiac events in the New York City area following the September 11, 2001, attack on the World Trade Center showed a 2.3-fold increase in the rate of tachyarrhythmias in patients with ICDs during the month after the attack (87), and another study showed a 68% increase in the frequency of defibrillator shocks for ventricular arrhythmias in ICD patients in Florida during the same month (88). Events less severe than the terrorist attacks also appear to increase cardiac events. In a 5-year retrospective study in England, mortality rates from myocardial infarction or stroke were increased by 1.28 times in men (but not in women) on days when the local professional soccer team lost (89). A study of over 200,000 Chinese and Japanese U.S. residents—ethnic groups that consider the number 4 to be particularly unlucky—found that cardiac mortality peaks on the fourth day of the month (90).

Chronic stress, such as job strain, also increases the risk of CHD. The Whitehall II study (91), a prospective cohort study of 6,895 male and 3,413 female civil servants 35–55 years of age followed over a period of 11 years on average, looked at rates of coronary death, angina, and nonfatal MI. Even after adjustment for age, sex, pay grade, and coronary risk factors, the employees with low decision latitude and high demands (high job strain) had an increased relative risk of CHD events compared with those without high job strain. The effect was most correlated with high job demands and less so with low decision latitude. Interestingly, the effect was more pronounced in lower age groups, and there was no effect difference related to employment grade or social support at work. The INTERHEART study, a recent very large multinational study of risk factors in myocardial infarction patients, revealed that stress levels were higher among individuals with new MI compared with age-, sex-, and site-matched control subjects. The population attributable risk for psychological stress, that is, the proportion of MI cases attributable to stress, was 12%–33% (92).

Anger, hostility, and type A behavior

Early research exploring the relationship of behavior patterns with cardiovascular disease focused on the effect of the type A behavior pattern (TABP)—time urgency, hostility, and achievement-striving-competitiveness—described in a seminal article by Friedman and Rosenman in 1959 (93). In 1975, the Western Collaborative Group Study of more than 3,000 middle-aged men found that over an 8-year period, men with type A behavior were twice as likely as men without those type A traits to develop coronary heart disease (94). Support of this theory was significantly weakened in 1985 by the publication of the MRFIT Behavior Pattern Study (95), a prospective study of 3,110 men with no history of a cardiac event. In the 7 years of follow-up there was no effect of TABP on the occurrence of MIs or coronary death. The Aspirin Myocardial Infarction Study prospectively followed 2,314 male and female heart attack survivors for at least 3 years. No correlation was found between the patients’ self-reported measures of type A behavior and the risk of MI or coronary death (96). A 1987 meta-analysis documented the weakening of the association of type A behavior with coronary heart disease outcomes in larger and newer studies (97). As a result, much of the subsequent research shifted to focus on specific traits within the type A construct, particularly hostility.

In 1980, Williams and colleagues studied 424 patients undergoing coronary angiography for suspected CHD, using both the TABP interview and the Cook-Medley Hostility Scale measurement, derived from the MMPI. Both TABP and Hostility Scale score were independently linked to the presence of atherosclerosis, but the Hostility Scale score was more strongly associated (98). A 25-year follow-up study of 255 physicians found that the Hostility Scale score was predictive of both CAD incidence and total mortality (99). Hostility Scale score has also been found to be associated with increased coronary artery calcification, a measure of atherosclerotic plaque development, in young asymptomatic men (100); with reduced high-frequency heart period variability, a risk factor for coronary and all-cause mortality, in young adults (101); and with increased platelet activation (102). These data stand in contrast to the finding that out of 14 population-based prospective cohort studies with at least 500 subjects published from 1974 to 1997, only six studies showed any link between TABP or hostility and CHD. If studies that include angina in their endpoint of cardiac events are excluded, only three of the 14 studies show any significant risk of cardiac events, with the relative risk ranging from 1.47 to 2.95 (103).

In patients with preexisting CHD, there is some evidence that hostility is associated with a worse prognosis. In 250 CHD patients and 500 control subjects who were followed over 8.5 years in the Western Collaborative Group Study, hostility was the only component independently linked to CHD incidence, with a relative risk of 1.93 (104). Some studies have found that in patients with existing CHD, hostility is associated with a higher risk of cardiac events, CHD mortality, and total mortality (105, 106). Hostility is also associated with more severe exercise-induced myocardial ischemia (107) and a higher risk of restenosis after percutaneous transluminal coronary angioplasty (PTCA) (108). In these studies there was significant variability in how hostility was measured.

There have been negative studies as well. A 33-year follow-up study of 1,399 men who completed the MMPI as freshmen at the University of Minnesota found that high Hostility Scale score did not predict CHD mortality or morbidity (109). In the Normative Aging Study, anger was found to predict the incidence of a combined endpoint (cardiac mortality, nonfatal MI, and angina) but did not significantly predict either cardiac mortality or nonfatal MI (110). Kaufmann and colleagues studied 331 post-MI patients and found that Hostility Scale score did not predict mortality at 6 or 12 months (10). One review identified five prospective cohort studies with 100 or more CHD patients that showed no prognostic role for type A or hostility measurements (103). Another study compared multiple questionnaire measures of anger and hostility in patients with CAD, in patients with valvular heart disease, and in normal control men. Only anger expression had a significant independent correlation with CAD severity (111). In summary, there may be a significant relationship between some aspect of anger and hostility and coronary disease incidence and prognosis, but it is clear that there is a lack of consensus on the best measure of anger and hostility as a cardiovascular risk factor. It is possible that self-report measures of anger and hostility are inevitably confounded by social desirability bias or lack of insight. Provocative data have been presented demonstrating that spouse ratings of men’s negative affects are superior to self-ratings of negative affects by male patients in their correlation with the men’s CAD risk (112).

Vital exhaustion

In European studies, vital exhaustion, defined as a state of diminished energy, increased irritability, and demoralization, has been identified as a coronary disease risk factor and a risk factor for coronary events and sudden cardiac arrest in patients with established CAD (113, 114). The overlap between vital exhaustion and depression has not been definitively determined, and few studies of the issue have been conducted in the United States. This concept requires further study.

Biological theories

A number of biological mechanisms may explain why patients with anxiety or depression have an elevated risk of CVD. Stress and depression cause dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. Under stress, the hypothalamus produces corticotropin-releasing factor (CRF), which stimulates the anterior pituitary to release corticotropin, signaling the adrenal glands to release cortisol. There is ample evidence of HPA hyperactivity in depressed patients, such as the presence of elevated levels of CRF in spinal fluid (115, 116) and nonsuppression of cortisol following administration of dexamethasone (117). Elevated levels of cortisol have been linked to the development of atherosclerosis and hypertension, although whether this is a direct or an indirect effect is unclear (118, 119). HPA hyperactivity is frequently accompanied by hyperactivity of the sympathoadrenal system. Chronic HPA and sympathoadrenal hyperactivity has been linked to atherosclerosis (120). Acute stress or anger initiates excessive release of epinephrine and norepinephrine, which elevates heart rate, increases blood pressure, and increases vascular resistance. Sympathetic hyperresponsiveness is also linked to the development of ischemia during exercise or mental stress (121, 122). Vagal withdrawal, that is, decreased parasympathetic tone, has been linked to mental stress, which can increase both heart rate and ventricular irritability.

Heart rate variability, which is controlled centrally by the hypothalamus, limbic system, and brainstem as well as peripherally by the vagus nerve, can be construed as an index of the ability to maintain cardiovascular homeostasis. In patients with cardiac disease, heart rate variability is decreased and is a predictor of cardiac mortality and morbidity (123125). Depression, hostility, and chronic worry are also linked to decreased heart rate variability (126129). This effect may partly explain the cardiovascular risks of depression and anxiety.

Anxiety disorders and stress cause ventricular instability, which would explain the link between anxiety disorders and sudden cardiac death rather than MI. Animals conditioned to anticipate unpleasant restraint will experience increased ventricular ectopy and ventricular tachycardia-fibrillation when exposed to the threat of restraint. This effect depends on the presence of current or past myocardial ischemia; stress alone does not induce events. In acute ischemia or vulnerable myocardium (e.g., previous infarction), stress induces ventricular tachycardia. The effect can be disrupted by the use of intracerebral β-adrenergic blockade (130136).

Depression (137141) and emotional stress (138, 142) are also linked to impairments in platelet function, such as increased reactivity and the secretion of platelet products, including platelet factor 4, β-thromboglobulin, and serotonin, which cause platelet aggregation. Platelet dysfunction can cause the formation of thrombi and also contribute to damage of vascular endothelial cells. Since selective serotonin reuptake inhibitors (SSRIs) reduce the ability of platelets to store serotonin, they may reduce the risk of thrombus formation (see the section Treatment Issues for Cardiac Patients). Chronic stress is also linked to alterations in the fibrinolytic system (143).

The link between chronic infection and inflammatory response and heart disease has been another area of interest in the past decade (144). Inflammatory responses such as increased leukocyte adhesiveness (145), T-cell activation (146, 147), and proinflammatory cytokines (148, 149) have been linked to depression and “burnout.” Ford and Erlinger (150) recently found that a history of depression was linked in a graduated fashion—based on temporal proximity of depression to the index measurement and number of episodes—to elevation of C-reactive protein in men.

Impact of psychiatric treatment on CVD

Can treatment of psychiatric conditions associated with CVD reduce the risk of developing CVD or improve the prognosis of patients who already have CVD? As the epidemiological findings of an association between psychiatric factors and heart disease have become more robust, these questions have taken on profound public health significance. Despite recent studies addressing the treatment of depression in patients with coronary disease, these questions remain largely unanswered (151, 152).

Medication interventions

Musselman and associates (153) studied the impact of paroxetine treatment on platelet reactivity in depressed patients. In this small nonrandomized study of 15 depressed and 12 nondepressed patients, none of whom had ischemic heart disease, the depressed patients had greater platelet activity and increased plasma concentrations of platelet factor 4. Treatment with paroxetine for 6 weeks in the depressed patients reduced all parameters of increased platelet activity. Confounding these results, 10 of the 15 depressed patients had one or more risk factors for ischemic heart disease (hypertension, obesity, and so on). None of the 12 control patients had any ischemic heart disease risk factors.

Sauer, Berlin, and Kimmel (154) conducted a case-control study of first MI in 653 smokers 30 to 65 years of age, compared with 2,990 community subjects as controls. After confounding variables were controlled for (including CVD risk factors, age, education, and physical activity), the odds ratio for MI among current subjects who used SSRIs within the index week was 0.35. There was a nonsignificant reduction in risk for those taking non-SSRI antidepressants, which was a very small group.

A large double-blind randomized controlled trial by Glassman and colleagues (152), the Sertraline Antidepressant Heart Attack Randomized Trial (SADHART), studied the safety and efficacy of sertraline treatment for 24 weeks in 369 patients initially hospitalized for acute MI or unstable angina. They reported a decreased incidence of “severe” cardiac events (as rated by the patients’ treating physicians) in the group treated with sertraline (14.5% compared with 22.4% on placebo), although the difference was not statistically significant (the study was not designed to have sufficient statistical power to test the significance of this effect). A follow-up study on the SADHART group data (155) showed that in a subset of 64 patients who participated in platelet studies, treatment with sertraline was associated with significantly decreased markers of platelet activation compared with placebo, an effect independent of the use of antiplatelet regimens, including aspirin, clopidogrel, or other antiplatelet agents and/or oral anticoagulants.

Sauer, Berlin, and Kimmel (156) conducted a case-control study of 1,080 first-MI patients 40 to 75 years of age and 4,256 control subjects who were followed over 3 years to determine the relationship of antidepressant serotonin transporter affinity to MI protection. After controlling for multiple CVD risk factors, SSRI users as a whole in this study did not have a significantly reduced risk of MI compared with nonusers. SSRI users taking high-affinity SSRIs (paroxetine, sertraline, and fluoxetine) did have a significantly reduced risk of MI compared with nonusers, with an odds ratio of 0.59. If confirmed by other studies, this result would suggest that high-affinity SSRIs might be of special benefit for patients at high risk of MI.

Electroconvulsive therapy

The effect of ECT on cardiac morbidity and mortality has not yet been adequately assessed. Schultz and colleagues examined nine patients with depression to test the hypothesis that treatment of depression with ECT would increase heart rate variability and potentially reduce the risk of cardiac mortality. Despite an improvement in patients’ depressive symptoms (mean Hamilton Depression Rating Scale scores decreased from 34.4 to 11.2), a decrease was noted in heart rate variability (157). In elderly patients with major depressive disorder, ECT responders have an increase in heart rate variability after treatment (158, 159).

Psychotherapy and supportive interventions

The Recurrent Coronary Prevention Project looked at 1,013 survivors of acute MI to see if group counseling to reduce type A behavior could reduce the risks of future MI and cardiac mortality (160). The patients were randomly assigned to a control condition, in which group cardiac counseling only was provided; an experimental condition, in which both group cardiac counseling and type A behavior counseling were provided; or a comparison group, which received no group counseling. The type A behavior counseling (primarily cognitive behavior psychotherapy techniques) was significantly more effective in reducing type A behavior than ordinary group counseling. The cumulative cardiac recurrence rate over 4.5 years was 12.9%, approximately half that in the group-cardiac-counseling-only group (21.2%) and in or the comparison group (28.2%).

Two large trials have failed to show any benefit of other specific psychosocial interventions on post-MI survival rates. In the Montreal Heart Attack Readjustment Trial (M-HART), 1,376 patients had monthly screenings for depression and anxiety as well as follow-up home nursing interventions for those who were “distressed.” Not only did the intervention have only a small impact on depression and anxiety, it had no impact on cardiac or all-cause mortality in men and showed a trend toward increasing cardiac and all-cause mortality in women (161). A multicenter study of 2,328 patients in England assessing a 7-week outpatient group program of psychological counseling, therapy, relaxation training, and stress management also found minimal impact on depression and anxiety and no impact on clinical sequelae or mortality (162). A 1996 meta-analysis of 23 randomized controlled trials evaluating the effects of psychosocial interventions on post-MI patients found that psychosocial treatment did reduce mortality and cardiac event recurrence rates over 2 years (163). Another meta-analysis of 37 psychoeducational programs (health education and stress management) for CHD patients, published in 1999, found a 34% decrease in cardiac mortality and a 29% decrease in recurrent MI (164).

The Enhancing Recovery in Coronary Heart Disease (ENRICHD) trial studied 2,481 patients from eight clinical centers within 28 days of their admission to the hospital for MI (151). Patients diagnosed with depression and/or low perceived social support were randomly assigned to a usual-care control group or to an intervention group. The intervention group received up to 6 months of individual cognitive behavior therapy sessions and group therapy for up to 3 additional months. After the initial treatment period, patients in either group could also take antidepressants if needed. No difference was seen in mortality or recurrent MI, even in the depressed subgroup of patients. Initially a modest but significantly greater improvement in depression and social support was observed in the treatment group, but by 42 months no difference could be seen. Notably, in patients on any antidepressant, and especially in the subgroup taking SSRIs, there was a trend toward decreased mortality but no difference in nonfatal MI. It should be noted that use of antidepressants was not randomized.

Treatment issues for cardiac patients

The safe psychopharmacological treatment of cardiac patients requires consideration of three main issues: (1) the cardioactive effects of psychiatric medications; (2) the interaction of psychotropic medications with other medications the patient is taking or is likely to be prescribed; (3) the impact of any comorbid health problems. All of these factors must be taken into account when deciding on which psychotropic medications to use in patients with cardiac disease.

Antidepressants

Except in the case of severe CHF causing reduced cardiac output, hepatic congestion, and renal impairment, the absorption, metabolism, and elimination of psychotropics other than lithium is generally not substantially impaired. Therefore, antidepressants must be used in therapeutically effective doses and not reduced unnecessarily for mild CHF.

Tricyclic antidepressants

Tricyclic antidepressants have many properties that make them relatively less desirable for use in cardiac patients. They cause cardiac conduction delays, including bundle-branch block or complete atrioventricular nodal block, and in overdose they may cause ventricular arrhythmias. Tricyclics are class IA antiarrhythmic agents, which prolong atrial and ventricular depolarization, causing increased P-R, QRS, and QT intervals. Studies have shown that QTc intervals of over 440 ms, and especially over 500 msec, are associated with an increased risk of sudden death (165, 166). Class IA antiarrhythmic agents have been proven to increase mortality in post-MI patients with premature ventricular contractions (167, 168).

Tricyclics can also cause significant orthostatic hypotension and tachycardia due to α1-adrenergic blockade. Patients with CHF are often on other medications that cause orthostatic hypotension, such as diuretics and vasodilators, and the risk of falling and sustaining fractures can be significant for elderly patients. Tachycardia increases cardiac demand in general and reduces left ventricular filling time, worsening diastolic function. Nortriptyline and desipramine tend to have fewer anticholinergic side effects and are better tolerated by cardiac patients than tertiary-amine tricyclics such as amitriptyline or doxepin (169171).

Given their effects, tricyclic antidepressants should be avoided whenever possible in patients with recent MI and should not be considered first-line agents for patients with ischemic heart disease or preexisting intraventricular conduction delays. In certain patients, the benefits may outweigh the risks, so consideration should be given to the entire clinical situation (172). When tricyclics are prescribed for patients with cardiac disease, orthostatic blood pressure measurements should be obtained at baseline and during treatment. A baseline ECG should also be obtained, and follow-up ECGs should be obtained when a therapeutic level of the drug has been reached to evaluate the P-R, QRS, and QTc intervals and to monitor for bundle-branch block or complete atrioventricular block (173).

Selective serotonin reuptake inhibitors

SSRIs have few cardiac effects in healthy patients. They can cause slowing of heart rate by a few beats per minute, which is usually clinically insignificant (174). A number of studies have confirmed that, overall, SSRIs carry little or no risk of increasing the QTc interval or causing other ECG changes (175177). Nonetheless, a few case reports have been published of significant sinus bradycardia, dysrhythmias, syncope (178), and QTc prolongation (179) in patients taking SSRIs.

In studies of patients who have preexisting cardiac disease, SSRIs have been found to have minimal negative effects on blood pressure or cardiac conduction. Until recently no randomized controlled trials with SSRIs and a placebo or an alternative treatment group with more than 81 total patients had been published (180, 181). The SADHART study, with 369 patients, was the first large randomized controlled trial to examine the effects of an SSRI, sertraline, in patients with depression after an acute coronary event. The study found that there was no effect of sertraline on heart rate, blood pressure, arrhythmias, ejection fraction, or cardiac conduction (152). Sertraline was found to be an effective treatment for depression, demonstrating a weak response in the overall sample but a very good response in the groups with severe and with recurrent major depressive disorder. Another small study showed a statistically significant 7% improvement in ejection fraction in patients with preexisting cardiac disease taking fluoxetine (182).

Cardiac effects of SSRIs may be dose dependent and vary from drug to drug. In a review of 6,000 ECGs of 1,789 patients treated with citalopram in clinical trials, no evidence was found of QTc prolongation (174). Studies of patients who have taken overdoses of various SSRIs indicate that in overdose citalopram causes significantly greater QTc prolongation than other SSRIs (183, 184). This may be related to the finding that citalopram inhibits cardiac sodium and calcium channels in animal studies (185). While very large doses (400 mg or more) were required to produce QTc prolongation in healthy patients, these data would suggest that, in general, extremely high doses of citalopram should be used with careful monitoring in cardiac patients, particularly those who are also taking inhibitors of cytochrome P450 enzyme 2C19 or 3A4.

Other antidepressants

Other antidepressants have not been as well studied with regard to cardiac effects. In a randomized double-blind crossover study of 10 patients with impaired left ventricular ejection fraction taking imipramine or bupropion, bupropion had no significant cardiac effects (171). In a study of 36 inpatients with cardiac disease, bupropion caused an increase in supine blood pressure, although it did not cause significant orthostatic hypotension, conduction disturbances, or ventricular arrhythmias. Bupropion did cause an exacerbation of baseline hypertension in two patients (186). Venlafaxine has not been specifically studied in cardiac patients, but it may cause a dose-dependent increase in diastolic blood pressure starting at 150 mg/day, with significant increases at doses of 300 mg/day or above (187). A 1-week randomized controlled trial of 20 patients—10 taking mirtazapine and 10 taking imipramine—found that mirtazapine caused a significant increase in heart rate and decrease in heart rate variability and had no effect on blood pressure or blood pressure variability (188).

Nefazodone is rarely used in cardiac patients because of multiple drug interactions. One small open-label study in CHF patients showed that it was effective in treating major depression, with a significant reduction in heart rate and no changes in heart rate variability. The QT interval was increased, which is consistent with a reduced heart rate, but the QTc interval did not change (189).

Monoamine oxidase inhibitors are almost never used in cardiac patients because of drug interactions, orthostatic hypotension, and the risk of hypertensive crises.

A number of SSRIs inhibit cytochrome P450 pathways, which has an impact on treatment decisions. Fluoxetine, paroxetine, and duloxetine inhibit 2D6. Nefazodone, fluoxetine, and, to a lesser extent, sertraline, are 3A4 inhibitors. Other medications, such as ketoconazole and erythromycin, are potent 3A4 inhibitors. Amiodarone and quinidine are 2D6 inhibitors and can elevate blood levels of fluoxetine, risperidone, and several tricyclics.

Tables 1, 2, and 3 summarize interactions between psychotropic, cardioactive, and cardiovascular drugs.

Antipsychotics

Antipsychotic medications may be used in small doses for short periods to treat delirium in cardiac patients who are in acute cardiac care settings or inpatient units. Longer-term and higher-dose therapy may be needed for cardiac patients who have a chronic psychotic disorder. The two situations are distinct, and some thought must be given to both the short-term and long-term risks and benefits of the various antipsychotic medications.

A number of antipsychotics have been linked to torsade de pointes and sudden death, among them pimozide, sertindole, droperidol, haloperidol, and thioridazine (190). The greatest risk is associated with thioridazine. In a large retrospective case-control study of all sudden deaths in psychiatric wards of five hospitals in England over 11 years, the only antipsychotic that was found to be an independent risk factor for sudden death was thioridazine (191). Of six major antipsychotics (thioridazine, ziprasidone, quetiapine, risperidone, olanzapine, and haloperidol), thioridazine produces the largest QTc prolongation, with a mean change of 35.6 ms (190, 192, 193). Several excellent reviews have examined the relationship of antipsychotics to QTc prolongation (191, 193198).

Among the atypical antipsychotics, none has been linked to torsade de pointes, even though most have a larger impact on QTc than haloperidol, which has been linked to torsade de pointes. Risperidone has been linked to one fatality due to pulseless electrical activity, and it has been reported to cause QTc and QRS prolongation in two cases of overdose and in 8 of 380 patients in a double-blind study by the manufacturer (199). The fatality may have been due to factors other than QTc prolongation (198). In two reported cases of ziprasidone overdose, neither patient developed torsade de pointes, though one had QRS prolongation and the other had QTc prolongation (198). Aripiprazole, olanzapine, and quetiapine have not been linked to torsade de pointes.

In considering the use of drugs that may prolong the QT interval, factors to be reviewed include a family or personal history of long QT syndrome, a history of sudden cardiac arrest, syncope or unexplained seizure, arrhythmias, hypertension, valvular heart disease, bradycardia, and use of other medications that may prolong the QT interval or interfere with the metabolism of QT-prolonging agents. Class IA and class III antiarrhythmic drugs, dolasetron, droperidol, tacrolimus, levomethadyl acetate, other antipsychotic agents, many antibiotics (the “floxacins”), and antifungal agents may increase the risk of torsade. Magnesium and potassium levels should be monitored, as abnormalities may also increase the vulnerability to developing torsade (190, 200, 201).

In addition to the potential proarrhythmic effects of the antipsychotics, the decision of which medication to select for patients with cardiac disease should take into account the drug’s potential to cause orthostatic hypotension, glucose intolerance, and hyperlipidemia. Orthostatic hypotension, which is related to the α1-adrenergic receptor blocking properties of antipsychotics, is seen most frequently with low-potency antipsychotics, such as chlorpromazine. Some atypical antipsychotics may also cause orthostatic hypotension. Hyperlipidemia, glucose intolerance, and diabetes mellitus, with or without weight gain, have been linked to most atypical antipsychotics, including clozapine, olanzapine, quetiapine, and risperidone. According to their manufacturers, aripiprazole and ziprasidone are less likely to cause these effects. In the initial phase of treatment, and periodically thereafter, blood sugar and lipid levels should be monitored.

Anxiolytics

Overall, benzodiazepines and buspirone are clinically safe and effective in patients with cardiac disease. Benzodiazepines have been shown to increase heart rate acutely, but they also reduce vagal tone and heart period variability, most likely by potentiating γ-aminobutyric acid (202, 203). Since they reduce anxiety and sympathetic nervous system activation, they decrease the heart rate and pressor responses to stress. On the other hand, buspirone, in one study, has been linked to decreased baseline heart rate, and it may actually enhance increased heart rate in response to stress (204).

Stimulants

Stimulants are particularly useful for treating apathy, fatigue, and psychomotor slowing in the medically ill, and they offer the advantage of working within a few days instead of several weeks. Dextroamphetamine and methylphenidate in doses of 5–30 mg/day are well tolerated by cardiac patients and have no significant effects on heart rate or blood pressure (205). They can also be used with good efficacy in poststroke patients, with no evidence of adverse cardiac effects (206).

Mood stabilizers

At therapeutic doses, lithium can cause sinus node dysfunction that is generally reversible when the medication is discontinued, although there have been rare reports of sinus arrest, atrioventricular block, and aggravation of ventricular arrhythmias (207209). Lithium toxicity can cause sinoatrial block, atrioventricular block or dissociation, bradyarrhythmias, and ventricular tachycardia or fibrillation (208). Generally, lithium can be used safely in cardiac patients even with reduced cardiac output by decreasing the dosage; the dosage can be decreased even further in patients with impaired renal function in advanced heart failure. Greater caution is necessary for patients taking diuretics, particularly thiazides, and those on salt-restricted diets. Lithium may also increase the risk of cardiac arrhythmias in patients taking angiotensin-converting enzyme inhibitors (194). In patients with acute congestive heart failure exacerbations or acute coronary syndromes, the use of lithium should generally be avoided because of rapid electrolyte and fluid balance shifts.

Valproate and lamotrigine have no apparent cardiovascular effects. Carbamazepine has a tricyclic-like class IA antiarrhythmic effect and should be used only with cautions similar to those discussed above. It also induces cytochrome P450 enzyme 3A4, which can increase the metabolism of a number of anticoagulant and cardiovascular medications.

Conclusion

Psychiatrists frequently treat patients with comorbid cardiovascular disease and can expect to do so increasingly with the aging of the population in the United States. Depression and anxiety are the most common problems arising in patients with heart disease, but patients with chronic psychotic illnesses and other mental disorders may also have heart disease. Fortunately, our knowledge base about treatment options is expanding. Whether the morbidity and mortality of cardiovascular disease can be modified through intervention aimed at psychiatric risk factors remains a research question of major importance.

Table 1. Impact of Psychotropic Medications on P450 Metabolism of Cardioactive Medications
Enzyme SubsystemCardioactive SubstratesPsychotropic InhibitorsPsychotropic Inducers
1A2PropranololFluvoxamineModafinil
 Verapamil  
 R-warfarin  
2B6No significant interactions  
2C19PropranololFluoxetineCarbamazepine
 R-warfarinFluvoxamine 
  Topiramate 
2C9Angiotensin II blockersFluvoxamineSecobarbital
  Sertraline 
 S-warfarin  
2D6Many anti-arrhythmics (encainide, flecainide, lidocaine)Bupropion 
  Clomipramine 
  Duloxetine 
  Fluoxetinea 
  Fluvoxamine 
 Many beta-blockersHaloperidol 
  Paroxetinea 
 CarvedilolSome phenothiazines 
3A4,5,7Calcium channel blockersNefazodoneaMany barbiturates
  Norfluoxetine (fluoxetine metabolite)Carbamazepine
 Lidocaine Modafinil
 Propranolol St. John’s wort
 Quinidine 3-OH (metabolite)Fluvoxamine 
 Many statins  

Source: Adapted from Flockhart (210)

a Particularly potent inhibitors

Table 1. Impact of Psychotropic Medications on P450 Metabolism of Cardioactive Medications
Enlarge table
Table 2. Impact of Cardioactive Medications on P450 Metabolism of Psychotropic Medications
Enzyme SubsystemPsychotropic SubstratesCardioactive InhibitorsCardioactive Inducers
1A2AmitriptylineTiclopidineNone
 Clomipramine  
 Olanzapine  
2B6BupropionTiclopidine 
2C19CitalopramNone 
 Diazepam (metabolite norphenytoin)  
 Some tricyclics  
2C9AmitriptylineAmiodarone 
 FluoxetineFluvastatin 
  Lovastatin 
2D6AmphetamineAmiodarone 
 Many antipsychotics (haloperidol, many phenothiazines, risperidone)Quinidine 
 Duloxetine  
 Fluoxetine  
 Fluvoxamine  
 Paroxetine  
 Many tricyclics  
 Venlafaxine  
3A4,5,7Many benzodiazepines (alprazolam, diazepam metabolite 3-OH, midazolam, triazolam)Amiodarone 
  Diltiazem 
  Verapamil 
 Buspirone  
 Carbamazepine  
 Haloperidol  
 Trazodone  
 Zaleplon  
 Zolpidem  
 Lidocaine  
 Propranolol  
 Quinidine 3-OH (metabolite)  
 Many statins  

Source: Adapted from Flockhart (210)

Table 2. Impact of Cardioactive Medications on P450 Metabolism of Psychotropic Medications
Enlarge table
Table 3. Selected Psychotropic Drug Interactions With Cardiovascular Drugs
Psychotropic AgentCardiovascular AgentEffect
SSRIsBeta-blockersAdditive bradycardic effects
SSRIsWarfarinIncreased bleeding risk, despite little effect on INR
Monoamine oxidase inhibitorsEpinephrine, dopamineHypertension
LithiumThiazide diureticsIncreased lithium level
Tricyclic antidepressantsGuanethidineReduced antihyper-tensive efficacy of guanethidine
Tricyclic anti-depressantsClass IA anti-arrhythmic agents, amiodaroneProlonged QT interval, increased atrioventricular block
LithiumAngiotensin-converting enzyme inhibitorsIncreased lithium level
PhenothiazinesBeta-blockersHypotension

INR=international normalized prothrombin ratio

Source: Shapiro (211)

Table 3. Selected Psychotropic Drug Interactions With Cardiovascular Drugs
Enlarge table

CME Disclosure Statement Julie K. Schulman, M.D., Department of Psychiatry, University of Colorado Health Sciences Center, Denver, Colorado.

Dr. Schulman reports no financial interests or affiliations with commercial organizations that may have direct or indirect interest in this CME program.

Philip R. Muskin, M.D., New York–Presbyterian Hospital—Columbia University Medical Center, New York, New York.

Consultant/speaker: Bristol-Myers Squibb, GlaxoSmithKline, Forest, Pfizer, AstraZeneca, Janssen, Cephalon. Research support: Bristol-Myers Squibb, Forest.

Peter A. Shapiro, M.D., Department of Psychiatry, Columbia University, New York, New York.

Speakers Bureau: Pfizer.

Acknowledgment Supported in part by the Nathaniel Wharton Fund (Dr. Shapiro).

Disclosure of Unapproved, Off-Label, or Investigational Use of a Product

APA policy requires disclosure by CME authors of unapproved or investigational use of products discussed in CME programs. Off-label use of medications by individual physicians is permitted and common. Decisions about off-label use can be guided by the scientific literature and clinical experience.

Send reprint requests to Dr. Muskin, Consultation-Liaison Psychiatry, New York–Presbyterian Hospital–Columbia University Medical Center, 622 West 168th Street, Box 427, New York, NY 10032.
References

1 American Heart Association: Heart Disease and Stroke Statistics: 2004 Update. Dallas, Tex, American Heart Association, 2003Google Scholar

2 American Psychiatric Association: Let’s Talk Facts About Anxiety Disorders. Washington, DC, American Psychiatric Association, 1999Google Scholar

3 American Psychiatric Association: Let’s Talk Facts About Depression. Washington, DC, American Psychiatric Association, 1998Google Scholar

4 Glassman AH, Shapiro PA: Depression and the course of coronary artery disease. Am J Psychiatry 1998; 155:4–11CrossrefGoogle Scholar

5 Rudisch B, Nemeroff CB: Epidemiology of comorbid coronary artery disease and depression. Biol Psychiatry 2003; 54:227–240CrossrefGoogle Scholar

6 Cannon WB: Voodoo death. Psychosom Med 1957; 19:182CrossrefGoogle Scholar

7 Schleifer SJ, Macari-Hinson MM, Coyle DA, Slater WR, Kahn M, Gorlin R, Zucker HD: The nature and course of depression following myocardial infarction. Arch Intern Med 1989; 149:1785–1789CrossrefGoogle Scholar

8 Forrester AW, Lipsey JR, Teitelbaum ML, DePaulo JR, Andrzejewski PL, Robinson RG: Depression following myocardial infarction. Int J Psychiatry Med 1992; 22:33–46CrossrefGoogle Scholar

9 Frasure-Smith N, Lesperance F, Talajic M: Depression following myocardial infarction: impact on 6-month survival. JAMA 1993; 270:1819–1825CrossrefGoogle Scholar

10 Kaufmann MW, Fitzgibbons JP, Sussman EJ, Reed JF, Einfalt JM, Rodgers JI, Fricchione GL: Relation between myocardial infarction, depression, hostility, and death. Am Heart J 1999; 138(3 Pt 1):549–554CrossrefGoogle Scholar

11 Luutonen S, Holm H, Salminen JK, Risla A, Salokangas RK: Inadequate treatment of depression after myocardial infarction. Acta Psychiatr Scand 2002; 106:434–439CrossrefGoogle Scholar

12 Lesperance F, Frasure-Smith N, Talajic M: Major depression before and after myocardial infarction: its nature and consequences. Psychosom Med 1996; 58:99–110CrossrefGoogle Scholar

13 Strik JJ, Lousberg R, Cheriex EC, Honig A: One year cumulative incidence of depression following myocardial infarction and impact on cardiac outcome. J Psychosom Res 2004; 56:59–66CrossrefGoogle Scholar

14 Aben I, Verhey F, Strik J, Lousberg R, Lodder J, Honig A: A comparative study into the one year cumulative incidence of depression after stroke and myocardial infarction. J Neurol Neurosurg Psychiatry 2003; 74:581–585CrossrefGoogle Scholar

15 Carney RM, Freedland KE, Sheline YI, Weiss ES: Depression and coronary heart disease: a review for cardiologists. Clin Cardiol 1997; 20:196–200CrossrefGoogle Scholar

16 Carney RM: Psychological risk factors for cardiac events: could there be just one? Circulation 1998; 97:128–129CrossrefGoogle Scholar

17 Hance M, Carney RM, Freedland KE, Skala J: Depression in patients with coronary heart disease: a 12-month follow-up. Gen Hosp Psychiatry 1996; 18:61–65CrossrefGoogle Scholar

18 Gonzalez MB, Snyderman TB, Colket JT, Arias RM, Jiang JW, O’Connor CM, Krishnan KR: Depression in patients with coronary artery disease. Depression 1996; 4:57–62CrossrefGoogle Scholar

19 Jiang W, Babyak MA, Rozanski A, Sherwood A, O’Connor CM, Waugh RA, Coleman RE, Hanson MW, Morris JJ, Blumenthal JA: Depression and increased myocardial ischemic activity in patients with ischemic heart disease. Am Heart J 2003; 146:55–61CrossrefGoogle Scholar

20 Blumenthal JA, Lett HS, Babyak MA, White W, Smith PK, Mark DB, Jones R, Matthew JP, Newman MF: Depression as a risk factor for mortality after coronary artery bypass surgery. Lancet 2003; 362(9384):604CrossrefGoogle Scholar

21 Connerney I, Shapiro PA, McLaughlin JS, Bagiella E, Sloan RP: Relation between depression after coronary artery bypass surgery and 12-month outcome: a prospective study. Lancet 2001; 358:1766–1771CrossrefGoogle Scholar

22 Shapiro PA, DePena M, Lidagoster L, Woodring S, Pierce DW, Glassman A: Depression after coronary artery bypass graft surgery. Psychosom Med 1998; 60:108Google Scholar

23 Freedland KE, Carney RM, Davila-Roman VG, Rich MW, Skala JA, Jaffe AS: Major depression and survival in congestive heart failure. Psychosom Med 1998; 60:118CrossrefGoogle Scholar

24 Jiang W, Alexander J, Christopher E, Kuchibhatla M, Gaulden LH, Cuffe MS, Blazing MA, Davenport C, Califf RM, Krishnan RR, O’Connor CM: Relationship of depression to increased risk of mortality and rehospitalization in patients with congestive heart failure. Arch Intern Med 2001; 161:1849–1856CrossrefGoogle Scholar

25 Faris R, Purcell H, Henein MY, Coats AJ: Clinical depression is common and significantly associated with reduced survival in patients with non-ischaemic heart failure. Eur J Heart Fail 2002; 4:541–551CrossrefGoogle Scholar

26 Moser DK, Dracup K: Is anxiety early after myocardial infarction associated with subsequent ischemic and arrhythmic events? Psychosom Med 1996; 58:395–401CrossrefGoogle Scholar

27 Cassem NH, Hackett TP: Psychiatric consultation in a coronary care unit. Ann Intern Med 1971; 75:9–14CrossrefGoogle Scholar

28 Sullivan M, LaCroix AZ, Spertus JA, Hecht J: Five-year prospective study of the effects of anxiety and depression on symptoms and function in patients with coronary heart disease. Am J Cardiol 2000; 86:1135–1138CrossrefGoogle Scholar

29 Bankier B, Januzzi JL, Littman AB: The high prevalence of multiple psychiatric disorders in stable outpatients with coronary heart disease. Psychosom Med 2004; 66:645–650CrossrefGoogle Scholar

30 Rafanelli C, Roncuzzi R, Finos L, Tossani E, Tomba E, Mangelli L, Urbinati S, Pinelli G, Fava GA: Psychological assessment in cardiac rehabilitation. Psychother Psychosom 2003; 72:343–349CrossrefGoogle Scholar

31 Lane D, Carroll D, Ring C, Beevers G, Lip GY: Mortality and quality of life 12 months after myocardial infarction: effects of depression and anxiety. Psychosom Med 2001; 63:221–230CrossrefGoogle Scholar

32 Rymaszewska J, Kiejna A, Hadrys T: Depression and anxiety in coronary artery bypass grafting patients. Eur Psychiatry 2003; 18:155–160CrossrefGoogle Scholar

33 MacMahon KMA, Lip GYH: Psychological factors in heart failure: a review of the literature. Arch Intern Med 2002; 162:509–516CrossrefGoogle Scholar

34 Riedinger MS, Dracup KA, Brecht M-L, for the SOLVD Investigators: Quality of life in women with heart failure, normative groups, and patients with other chronic conditions. Am J Crit Care 2002; 11:211–219Google Scholar

35 Steptoe A, Mohabir A, Mahon NG, McKenna WJ: Health related quality of life and psychological wellbeing in patients with dilated cardiomyopathy. Heart 2000; 83:645–650CrossrefGoogle Scholar

36 DiMarco JP: Implantable cardioverter-defibrillators. N Engl J Med 2003; 349:1836–1847CrossrefGoogle Scholar

37 Hamner M, Hunt N, Gee J, Garrell R, Monroe R: PTSD and automatic implantable cardioverter defibrillators. Psychosomatics 1999; 40:82–85CrossrefGoogle Scholar

38 Morris PL, Badger J, Chmielewski C, Berger E, Goldberg RJ: Psychiatric morbidity following implantation of the automatic implantable cardioverter defibrillator. Psychosomatics 1991; 32:58–64CrossrefGoogle Scholar

39 Chevalier P, Verrier P, Kirkorian G, Touboul P, Cottraux J: Improved appraisal of the quality of life in patients with automatic implantable cardioverter defibrillator: a psychometric study. Psychother Psychosom 1996; 65:49–56CrossrefGoogle Scholar

40 Crow SJ, Collins J, Justic M, Goetz R, Adler S: Psychopathology following cardioverter defibrillator implantation. Psychosomatics 1998; 39:305–310CrossrefGoogle Scholar

41 Bourke JP, Turkington D, Thomas G, McComb JM, Tynan M: Florid psychopathology in patients receiving shocks from implanted cardioverter-defibrillators. Heart 1997; 78:581–583CrossrefGoogle Scholar

42 Godemann F, Ahrens B, Behrens S, Berthold R, Gandor C, Lampe F, Linden M: Classic conditioning and dysfunctional cognitions in patients with panic disorder and agoraphobia treated with an implantable cardioverter/defibrillator. Psychosom Med 2001; 63:231–238CrossrefGoogle Scholar

43 Herrmann C, von zur Muhen F, Schaumann A, Buss U, Kemper S, Wantzen C, Gonska BD: Standardized assessment of psychological well-being and quality-of life in patients with implanted defibrillators. Pacing Clin Electrophysiol 1997; 20(1 Pt 1):95–103CrossrefGoogle Scholar

44 Heller SS, Ormont MA, Lidagoster L, Sciacca RR, Steinberg S: Psychosocial outcome after ICD implantation: a current perspective. Pacing Clin Electrophysiol 1998; 21:1207–1215CrossrefGoogle Scholar

45 Newman DM, Dorian P, Paquette M, Sulke N, Gold MR, Schwartzman DS, Schaaf K, Wood K, Johnson L: Effect of an implantable cardioverter defibrillator with atrial detection and shock therapies on patient-perceived, health-related quality of life. Am Heart J 2003; 145:841–846CrossrefGoogle Scholar

46 Duru F, Buchi S, Klaghofer R, Mattmann H, Sensky T, Buddeberg C, Candinas R: How different from pacemaker patients are recipients of implantable cardioverter-defibrillators with respect to psychosocial adaptation, affective disorders, and quality of life? Heart 2001; 85:375–379CrossrefGoogle Scholar

47 INTERHEART Study Investigators: Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet 2004; 364(9438): 937–952CrossrefGoogle Scholar

48 Fergusson DM, Goodwin RD, Horwood LJ: Major depression and cigarette smoking: results of a 21-year longitudinal study. Psychol Med 2003; 33:1357–1367CrossrefGoogle Scholar

49 Breslau N, Peterson EL, Schultz LR, Chilcoat HD, Andreski P: Major depression and stages of smoking: a longitudinal investigation. Arch Gen Psychiatry 1998;55:161–166CrossrefGoogle Scholar

50 Hall SM, Munoz RF, Reus VI, Sees KL: Nicotine, negative affect, and depression. J Consult Clin Psychol 1993; 61:761–767CrossrefGoogle Scholar

51 Anda RF, Williamson DF, Escobedo LG, Mast EE, Giovino GA, Remington PL: Depression and the dynamics of smoking. JAMA 1990; 264:1541–1545CrossrefGoogle Scholar

52 Quattrocki E, Baird A, Yurgelun-Todd D: Biological aspects of the link between smoking and depression. Harv Rev Psychiatry 2000; 8:99–110CrossrefGoogle Scholar

53 McChargue DE, Spring B, Cook JW, Neumann CA: Reinforcement expectations explain the relationship between depressive history and smoking status in college students. Addict Behav 2004; 29:991–994CrossrefGoogle Scholar

54 John U, Meyer C, Rumpf HJ, Hapke U: Self-efficacy to refrain from smoking predicted by major depression and nicotine dependence. Addict Behav 2004; 29:857–866CrossrefGoogle Scholar

55 Garcia MJ, McNamara PM, Gordon T, Kannel WB: Morbidity and mortality in diabetics in the Framingham population sixteen year follow-up study. Diabetes 1974; 23:105–111CrossrefGoogle Scholar

56 Kawakami N, Takatsuka N, Shimizu H, Ishibashi H: Depressive symptoms and occurrence of type 2 diabetes among Japanese men. Diabetes Care 1999; 22:1071–1076CrossrefGoogle Scholar

57 Golden SH, Williams JE, Ford DE, Yeh H-C, Paton Sanford C, Nieto FJ, Brancati FL: Depressive symptoms and the risk of type 2 diabetes: the Atherosclerosis Risk in Communities study. Diabetes Care 2004; 27:429–435CrossrefGoogle Scholar

58 Arroyo C, Hu FB, Ryan LM, Kawachi I, Colditz GA, Speizer FE, Manson J: Depressive symptoms and risk of type 2 diabetes in women. Diabetes Care 2004; 27:129–133CrossrefGoogle Scholar

59 Pine DS, Goldstein RB, Wolk S, Weissman MM: The association between childhood depression and adulthood body mass index. Pediatrics 2001; 107:1049–1056CrossrefGoogle Scholar

60 Goodman E, Whitaker RC: A prospective study of the role of depression in the development and persistence of adolescent obesity. Pediatrics 2002; 110:497–504CrossrefGoogle Scholar

61 Musselman DL, Evans DL, Nemeroff CB: The relationship of depression to cardiovascular disease: epidemiology, biology, and treatment. Arch Gen Psychiatry 1998; 55:580–592CrossrefGoogle Scholar

62 Rugulies R: Depression as a predictor for coronary heart disease: a review and meta-analysis. Am J Prev Med 2002; 23:51–61CrossrefGoogle Scholar

63 Aromaa A, Raitasalo R, Reunanen A, Impivaara O, Heliovaara M, Knekt P, Lehtinen V, Joukamaa M, Maatela J: Depression and cardiovascular diseases. Acta Psychiatr Scand 1994; 377(suppl):77–82Google Scholar

64 Pratt LA, Ford DE, Crum RM, Armenian HK, Gallo JJ, Eaton WW: Depression, psychotropic medication, and risk of myocardial infarction: prospective data from the Baltimore ECA follow-up. Circulation 1996; 94:3123–3129CrossrefGoogle Scholar

65 Penninx BWJH, Beekman ATF, Honig A, Deeg DJH, Schoevers RA, van Eijk JTM, van Tilburg W: Depression and cardiac mortality: results from a community-based longitudinal study. Arch Gen Psychiatry 2001; 58:221–227CrossrefGoogle Scholar

66 Barefoot JC, Brummett BH, Helms MJ, Mark DB, Siegler IC, Williams RB: Depressive symptoms and survival of patients with coronary artery disease. Psychosom Med 2000; 62:790–795CrossrefGoogle Scholar

67 Carney RM, Rich MW, Freedland KE, Saini J, teVelde A, Simeone C, Clark K: Major depressive disorder predicts cardiac events in patients with coronary artery disease. Psychosom Med 1988; 50:627–633CrossrefGoogle Scholar

68 Burg MM, Benedetto MC, Soufer R: Depressive symptoms and mortality two years after coronary artery bypass graft surgery (CABG) in men. Psychosom Med 2003; 65:508–510CrossrefGoogle Scholar

69 Borowicz L Jr, Royall R, Grega M, Selnes O, Lyketsos C, McKhann G: Depression and cardiac morbidity 5 years after coronary artery bypass surgery. Psychosomatics 2002; 43:464–471CrossrefGoogle Scholar

70 Frasure-Smith N, Lesperance F, Talajic M: Depression and 18-month prognosis after myocardial infarction. Circulation 1995; 91:999–1005CrossrefGoogle Scholar

71 Lesperance F, Frasure-Smith N, Talajic M, Bourassa MG: Five-year risk of cardiac mortality in relation to initial severity and one-year changes in depression symptoms after myocardial infarction. Circulation 2002; 105:1049–1053CrossrefGoogle Scholar

72 Irvine J, Basinski A, Baker B, Jandciu S, Paquette M, Cairns J, Connolly S, Roberts R, Gent M, Dorian P: Depression and risk of sudden cardiac death after acute myocardial infarction: testing for the confounding effects of fatigue. Psychosom Med 1999; 61:729–737CrossrefGoogle Scholar

73 Strik JJ, Denollet J, Lousberg R, Honig A: Comparing symptoms of depression and anxiety as predictors of cardiac events and increased health care consumption after myocardial infarction. J Am Coll Cardiol 2003; 42:1801–1807CrossrefGoogle Scholar

74 Ladwig KH, Kieser M, Konig J, Breithardt G, Borggrefe M: Affective disorders and survival after acute myocardial infarction: results from the post-infarction late potential study. Eur Heart J 1991; 12:959–964Google Scholar

75 Murberg TA, Bru E, Svebak S, Tveteras R, Aarsland T: Depressed mood and subjective health symptoms as predictors of mortality in patients with congestive heart failure: a two-year follow-up study. Int J Psychiatry Med 1999; 29:311–326CrossrefGoogle Scholar

76 Hedayati SS, Jiang W, O’Connor CM, Kuchibhatla M, Krishnan KR, Cuffe MS, Blazing MA, Szczech LA: The association between depression and chronic kidney disease and mortality among patients hospitalized with congestive heart failure. Am J Kidney Dis 2004; 44:207–215CrossrefGoogle Scholar

77 Krumholz HM, Butler J, Miller J, Vaccarino V, Williams CS, Mendes de Leon CF, Seeman TE, Kasl SV, Berkman LF: Prognostic importance of emotional support for elderly patients hospitalized with heart failure. Circulation 1998; 97:958–964CrossrefGoogle Scholar

78 Koenig HG: Depression in hospitalized older patients with congestive heart failure. Gen Hosp Psychiatry 1998; 20:29–43CrossrefGoogle Scholar

79 Haines AP, Imeson JD, Meade TW: Phobic anxiety and ischemic heart disease. BMJ 1987; 295:297–299CrossrefGoogle Scholar

80 Kawachi I, Colditz G, Ascherio A, Rimm E, Giovannucci E, Stampfer M, Willett W: Prospective study of phobic anxiety and risk of coronary heart disease in men. Circulation 1994; 89:1992–1997CrossrefGoogle Scholar

81 Kawachi I, Sparrow D, Vokonas PS, Weiss ST: Symptoms of anxiety and risk of coronary heart disease: the normative aging study. Circulation 1994; 90:2225–2229CrossrefGoogle Scholar

82 Frasure-Smith N, Lesperance F, Talajic M: The impact of negative emotions on prognosis following myocardial infarction: is it more than depression? Health Psychology 1995; 14:388–398CrossrefGoogle Scholar

83 Denollet J, Brutsaert DL: Personality, disease severity, and the risk of long term cardiac events in patients with a decreased ejection fraction after myocardial infarction. Circulation 1998; 97:167–173CrossrefGoogle Scholar

84 Kaprio J, Koskenvuo M, Rita H: Mortality after bereavement: a prospective study of 95,647 persons. Am J Public Health 1987; 77:283–287CrossrefGoogle Scholar

85 Leor J, Kloner RA: The Northridge earthquake as a trigger for acute myocardial infarction. Am J Cardiol 1996; 77:1230–1232CrossrefGoogle Scholar

86 Leor J, Poole WK, Kloner RA: Sudden cardiac death triggered by an earthquake. N Engl J Med 1996; 334:413–419CrossrefGoogle Scholar

87 Steinberg JS, Arshad A, Kowalski M, Kukar A, Suma V, Vloka M, Ehlert F, Herweg B, Donnelly J, Philip J, Reed G, Rozanski A: Increased incidence of life-threatening ventricular arrhythmias in implantable defibrillator patients after the World Trade Center attack. J Am Coll Cardiol 2004; 44:1261–1264CrossrefGoogle Scholar

88 Shedd OL, Sears SF Jr, Harvill JL, Arshad A, Conti JB, Steinberg JS, Curtis AB: The World Trade Center attack: increased frequency of defibrillator shocks for ventricular arrhythmias in patients living remotely from New York City. J Am Coll Cardiol 2004; 44:1265–1267CrossrefGoogle Scholar

89 Kirkup W, Merrick DW: A matter of life and death: population mortality and football results. J Epidemiol Community Health 2003; 57:429–432CrossrefGoogle Scholar

90 Phillips DP, Liu GC, Kwok K, Jarvinen JR, Zhang W, Abramson IS: The Hound of the Baskervilles effect: natural experiment on the influence of psychological stress on timing of death. BMJ 2001; 323(7327): 1443–1446CrossrefGoogle Scholar

91 Kuper H, Marmot M: Job strain, job demands, decision latitude, and risk of coronary heart disease within the Whitehall II study. J Epidemiol Community Health 2003; 57:147–153CrossrefGoogle Scholar

92 Rosengren A, Hawken S, Ounpuu S, Sliwa K, Zubaid M, Almahmeed WA, Blackett KN, Sitthiamorn C, Sato H, Yusuf S; INTERHEART investigators: Association of psychosocial risk factors with risk of acute myocardial infarction in 11119 cases and 13648 controls from 52 countries (the INTERHEART study): case-control study. Lancet 2004; 364(9438):953–962CrossrefGoogle Scholar

93 Friedman M, Rosenman RH: Association of specific overt behavior pattern with blood and cardiovascular findings: blood cholesterol level, blood clotting time, incidence of arcus senilis, and clinical coronary artery disease. JAMA 1959; 169:1286–1296CrossrefGoogle Scholar

94 Rosenman RH, Brand RJ, Jenkins CD, Friedman M, Straus R, Wurm M: Coronary heart disease in the Western Collaborative Group Study: final follow-up experience of 8 1/2 years. JAMA 1975; 233:872–877CrossrefGoogle Scholar

95 Shekelle RB, Hulley SB, Neaton JD, Billings JH, Borhani NO, Gerace TA, Jacobs DR, Lasser NL, Mittlemark MB, Stamler J: The MRFIT Behavior Pattern Study, II: type A behavior and incidence of coronary heart disease. Am J Epidemiol 1985; 122:559–570CrossrefGoogle Scholar

96 Shekelle RB, Gale M, Norusis M: Type A score (Jenkins Activity Survey) and risk of recurrent coronary heart disease in the Aspirin Myocardial Infarction Study. Am J Cardiol 1985; 56:221–225Google Scholar

97 Booth-Kewley S, Friedman HS: Psychological predictors of heart disease: a quantitative review. Psychol Bull 1987; 101:343–362CrossrefGoogle Scholar

98 Williams RB Jr, Haney TL, Lee KL, Kong YH, Blumenthal JA, Whalen RE: Type A behavior, hostility, and coronary atherosclerosis. Psychosom Med 1980; 42:539–549CrossrefGoogle Scholar

99 Barefoot J, Dahlstrom WG, Williams RB: Hostility, CHD incidence, and total mortality: a 25-year follow-up study of 255 physicians. Psychosom Med 1983; 45:59–63CrossrefGoogle Scholar

100 Iribarren C, Sidney S, Bild DE, Liu K, Markovitz JH, Roseman JM, Matthews K: Association of hostility with coronary artery calcification in young adults: the CARDIA study (Coronary Artery Risk Development in Young Adults). JAMA 2000; 283:2546–2551CrossrefGoogle Scholar

101 Sloan RP, Shapiro PA, Bigger T Jr, Bagiella E, Steinman RC, Gorman JM: Cardiac autonomic control and hostility in healthy subjects. Am J Cardiol 1994; 74:298–300CrossrefGoogle Scholar

102 Markovitz JH: Hostility is associated with increased platelet activation in coronary heart disease. Psychosom Med 1998; 60:586–591CrossrefGoogle Scholar

103 Hemingway H, Marmot M: Evidence based cardiology: psychosocial factors in the aetiology and prognosis of coronary heart disease: systematic review of prospective cohort studies. BMJ 1999; 318(7196):1460–1467CrossrefGoogle Scholar

104 Hecker MHL, Chesney MA, Blacks GW, Frautschi N: Coronary-prone behaviors in the Western Collaborative Group Study. Psychosom Med 1988; 50:153–164CrossrefGoogle Scholar

105 Boyle SH, Williams RB, Mark DB, Brummett BH, Siegler IC, Helms MJ, Barefoot JC: Hostility as a predictor of survival in patients with coronary artery disease. Psychosom Medicine 2004; 66:629–632CrossrefGoogle Scholar

106 Dembroski TM, MacDougall JM, Costa PT, Grandits GA: Components of hostility as predictors of sudden death and myocardial infarction in the Multiple Risk Factor Intervention Trial. Psychosom Med 1989; 51:514–522CrossrefGoogle Scholar

107 Helmers KF, Krantz DS, Howell RH, Klein J, Bairey N, Rozanski A: Hostility and myocardial ischemia in coronary artery disease patients: evaluation by gender and ischemic index. Psychosom Med 1993; 55:29–36CrossrefGoogle Scholar

108 Goodman M, Quigley J, Moran G, Meilman H, Sherman M: Hostility predicts restenosis after percutaneous transluminal coronary angioplasty. Mayo Clin Proc 1996; 71:729–734CrossrefGoogle Scholar

109 Hearn MD, Murray DM, Luepker RV: Hostility, coronary heart disease, and total mortality: a 33-year follow-up study of university students. J Behav Med 1989; 12:105–121CrossrefGoogle Scholar

110 Kawachi I, Sparrow D, Spiro A 3rd, Vokonas P, Weiss ST: A prospective study of anger and coronary heart disease: the Normative Aging Study. Circulation 1996; 94:2090–2095CrossrefGoogle Scholar

111 McDermott MR, Ramsay JM, Bray C: Components of the anger-hostility complex as risk factors for coronary artery disease severity: a multi-measure study. J Health Psychol 2001; 6:309–319CrossrefGoogle Scholar

112 Ketterer M, Denollet J, Chapp J, Thayer B, Keteyian S, Clark V, John S, Farha A, Deveshwar S: Men deny and women cry, but who dies? Do the wages of “denial” include early ischemic coronary heart disease? J Psychosom Res 2004; 56:119–123CrossrefGoogle Scholar

113 Appels A, Mulder P: Excess fatigue as a precursor of myocardial infarction. Eur Heart J 1988; 9:758–764CrossrefGoogle Scholar

114 Appels A, Mulder P: Fatigue and heart disease: the association between “vital exhaustion” and past, present, and future coronary heart disease. J Psychosom Res 1989; 3:727–738CrossrefGoogle Scholar

115 Nemeroff CB, Widerlov E, Bissette G, Walleus H, Karisson I, Eklund K, Kitts CD, Loosen PT, Vale W: Elevated concentrations of CSF corticotropin releasing factor-like immunoreactivity in depressed patients. Science 1984; 226:1342–1344CrossrefGoogle Scholar

116 Banki C, Bissette G, Arato M, O’Connor L, Nemeroff C: CSF corticotropin-releasing factor-like immunoreactivity in depression and schizophrenia. Am J Psychiatry 1987; 144:873–877CrossrefGoogle Scholar

117 Coryell W, Schlesser M: The dexamethasone suppression test and suicide prediction. Am J Psychiatry 2001; 158:748–753CrossrefGoogle Scholar

118 Colao A, Pivonello R, Spiezia S, Faggiano A, Ferone D, Filippella M, Marzullo P, Cerbone G, Siciliani M, Lombardi G: Persistence of increased cardiovascular risk in patients with Cushing’s disease after five years of successful cure. J Clin Endocrinol Metab 1999; 84:2664–2672Google Scholar

119 Troxler RG, Sprague EA, Albanese RA, Fuchs R, Thompson AJ: The association of elevated plasma cortisol and early atherosclerosis as demonstrated by coronary angiography. Atherosclerosis 1977; 26:151–162CrossrefGoogle Scholar

120 Kaplan JR, Pettersson K, Manuck SB, Olsson G: Role of sympathoadrenal medullary activation in the initiation and progression of atherosclerosis. Circulation 1991;8 4(suppl VI):VI-23–VI-32Google Scholar

121 Goldberg AD, Becker LC, Bonsall R, Cohen JD, Ketterer MW, Kaufman PG, Krantz DS, Light KC, McMahon RP, Noreuil T, Pepine CJ, Raczynski J, Stone PH, Strother D, Taylor H, Sheps DS: Ischemic, hemodynamic, and neurohormonal responses to mental and exercise stress: experience from the psychophysiological investigations of myocardial ischemia study (PIMI). Circulation 1996; 94:2402–2409CrossrefGoogle Scholar

122 Krantz DS, Helmers KF, Bairey CN, Nebel LE, Hedges SM, Rosanski A: Cardiovascular reactivity and mental stress-induced myocardial ischemia in patients with coronary artery disease. Psychosom Med 1991; 53:1–12CrossrefGoogle Scholar

123 Farrell TG, Bashir Y, Cripps TR, Malik M, Poloniecki J, Bennett ED, Ward D, Camm AJ: Heart rate variability and sudden death secondary to coronary artery disease during ambulatory electrocardiographic monitoring. Am J Cardiol 1987; 60:86–89CrossrefGoogle Scholar

124 Huikuri HV, Makikallio TH: Heart rate variability in ischemic heart disease. Auton Neurosci 2001; 90:95–101CrossrefGoogle Scholar

125 Kleiger RE, Miller JP, Bigger JT Jr, Moss AJ: Decreased heart rate variability and its association with increased mortality after acute myocardial infarction. Am J Cardiol 1987; 59:256–262CrossrefGoogle Scholar

126 Carney RM, Blumenthal JA, Stein PK, Watkins L, Catellier D, Berkman LF, Czajkowski SM, O’Connor C, Stone PH, Freedland KE: Depression, heart rate variability, and acute myocardial infarction. Circulation 2001; 104:2024–2028CrossrefGoogle Scholar

127 Kubzansky LD, Kawachi I, Spiro A III, Weiss ST, Vokonas PS, Sparrow D: Is worrying bad for your heart? A prospective study of worry and coronary heart disease in the normative aging study. Circulation 1997; 95:818–824CrossrefGoogle Scholar

128 Carney RM, Rich MW, teVelde A, Saini J, Clark K, Freedland KE: The relationship between heart rate, heart rate variability, and depression in patients with coronary artery disease. J Psychosom Res 1988; 32:159–164CrossrefGoogle Scholar

129 Stein PK, Carney RM, Freedland KE, Skala JA, Jaffe AS, Kleiger RE, Rottman JN: Severe depression is associated with markedly reduced heart rate variability in patients with stable coronary heart disease. J Psychosom Res 2000; 48:493–500CrossrefGoogle Scholar

130 Lown B, DeSilva RA, Reich P, Murawski BJ: Psychophysiologic factors in sudden cardiac death. Am J Psychiatry 1980; 137:1325–1335CrossrefGoogle Scholar

131 Lown B: Role of higher nervous activity in sudden cardiac death. Jpn Circ J 1990; 54:581–602CrossrefGoogle Scholar

132 DeSilva RA: Central nervous system risk factors for sudden cardiac death. J S C Med Assoc 1983; 79:561–572Google Scholar

133 DeSilva RA: Cardiac arrhythmias and sudden cardiac death, in Medical-Psychiatric Practice. Edited by Stoudemire A, Fogel BS. Washington, DC, American Psychiatric Press, 1993, pp 199–236Google Scholar

134 Skinner JE, Lie JT, Entman ML: Modification of ventricular fibrillation latency following coronary artery occlusion in the conscious pig. Circulation 1975; 51:656–667CrossrefGoogle Scholar

135 Skinner JE, Reed JC: Blockade of a frontocortical-brainstem pathway prevents ventricular fibrillation of the ischemic heart in pigs. Am J Physiol 1981; 240:H156–H163Google Scholar

136 Skinner JE: Regulation of cardiac vulnerability by the cerebral defense system. J Am Coll Cardiol 1985; 5:88B–94BCrossrefGoogle Scholar

137 Fitzgerald DJ, Roy L, Catella F, FitzGerald GA: Platelet activation in unstable coronary disease. N Engl J Med 1986; 315:983–989CrossrefGoogle Scholar

138 Grignani G, Soffiantino F, Zucchella M, Pacchiarini L, Tacconi F, Bonomi E, Pastoris A, Sbaffi A, Fratino P, Tavazzi L: Platelet activation by emotional stress in patients with coronary artery disease. Circulation 1991; 83(suppl II):II-128–II-136Google Scholar

139 Musselman DL, Tomer A, Manatunga AK, Knight BT, Porter MR, Kasey S, Marzec U, Harker LA, Nemeroff CB: Exaggerated platelet reactivity in major depression. Am J Psychiatry 1996; 153:1313–1317CrossrefGoogle Scholar

140 Kuijpers PM, Hamulyak K, Strik JJ, Wellens HJ, Honig A: Beta-thromboglobulin and platelet factor 4 levels in post-myocardial infarction patients with major depression. Psychiatry Res 2002; 109:207–210CrossrefGoogle Scholar

141 Shimbo D, Child J, Davidson K, Geer E, Osende JI, Reddy S, Dronge A, Fuster V, Badimon JJ: Exaggerated serotonin-mediated platelet reactivity as a possible link in depression and acute coronary syndromes. Am J Cardiol 2002; 89:331–333CrossrefGoogle Scholar

142 Levine SP, Towell BL, Suarez AM, Knieriem LK, Harris MM, George JN: Platelet activation and secretion associated with emotional stress. Circulation 1985; 71:1129–1134CrossrefGoogle Scholar

143 Raikkonen K, Lassila R, Keltikangas-Jarvinen L, Hautanen A: Association of chronic stress with plasminogen activator inhibitor-1 in healthy middle-aged men. Arterioscler Thromb Vasc Biol 1996; 16:363–367CrossrefGoogle Scholar

144 Danesh J, Collins R, Peto R: Chronic infections and coronary heart disease: is there a link? Lancet 1997: 350:430–436CrossrefGoogle Scholar

145 Lerman Y, Melamed S, Shragin Y, Kushnir T, Rotgoltz Y, Shirom A, Aronson M: Association between burnout at work and leukocyte adhesiveness/aggregation. Psychosom Med 1999; 61:828–833CrossrefGoogle Scholar

146 Maes M, Van der Planken M, Stevens WJ, Peeters D, DeClerck LS, Bridts CH, Schotte C, Cosyns P: Leukocytosis, monocytosis, and neutrophilia: hallmarks of severe depression. J Psychiatr Res 1992; 26:125–134CrossrefGoogle Scholar

147 Maes M, Stevens WJ, Declerck LS, Bridts CH, Peeters D, Schotte C, Cosyns P: Significantly increased expression of T-cell activation markers (interleukin-2 and HLA-DR) in depression: further evidence for an inflammatory process during that illness. Prog Neuropsychopharmacol Biol Psychiatry 1993; 17:241–255CrossrefGoogle Scholar

148 Licinio J, Wong ML: The role of inflammatory mediators in the biology of major depression: central nervous system cytokines modulate the biological substrate of depressive symptoms, regulate stress-responsive systems, and contribute to neurotoxicity and neuroprotection. Mol Psychiatry 1999; 4:317–327CrossrefGoogle Scholar

149 Appels A, Bar FW, Bar J, Bruggeman C, de Baets M: Inflammation, depressive symptomatology, and coronary artery disease. Psychosom Med 2000; 62:601–605CrossrefGoogle Scholar

150 Ford DE, Erlinger TP: Depression and C-reactive protein in US adults: data from the Third National Health and Nutrition Examination Survey. Arch Intern Med 2004; 164:1010–1014CrossrefGoogle Scholar

151 Writing Committee for the ENRICHD Investigators: Effects of treating depression and low perceived social support on clinical events after myocardial infarction: the Enhancing Recovery in Coronary Heart Disease (ENRICHD) randomized trial. JAMA 2003; 289:3106–3116CrossrefGoogle Scholar

152 Glassman AH, O’Connor CM, Califf RM, Swedberg K, Schwartz P, Bigger JT Jr, Krishnan KR, van Zyl LT, Swenson JR, Finkel MS, Landau C, Shapiro PA, Pepine CJ, Mardekian J, Harrison WM, Barton D, Mclvor M; Sertraline Antidepressant Heart Attack Randomized Trial (SADHEART) Group: Sertraline treatment of major depression in patients with acute MI or unstable angina. JAMA 2002; 288:701–709CrossrefGoogle Scholar

153 Musselman DL, Marzec UM, Manatunga A, Penna S, Reemsnyder A, Knight BT, Baron A, Hanson SR, Nemeroff CB: Platelet reactivity in depressed patients treated with paroxetine: preliminary findings. Arch Gen Psychiatry 2000; 57:875–882CrossrefGoogle Scholar

154 Sauer WH, Berlin JA, Kimmel SE: Selective serotonin reuptake inhibitors and myocardial infarction. Circulation 2001; 104:1894–1898CrossrefGoogle Scholar

155 Serebruany VL, Glassman AH, Malinin AI, Nemeroff CB, Musselman DL, van Zyl LT, Finkel MS, Krishnan KR, Gaffney M, Harrison W, Califf RM, O’Connor CM: Platelet/endothelial biomarkers in depressed patients treated with the selective serotonin reuptake inhibitor sertraline after acute coronary events: the Sertraline AntiDepressant Heart Attack Randomized Trial (SADHART) Platelet Substudy. Circulation 2003; 108:939–944CrossrefGoogle Scholar

156 Sauer WH, Berlin JA, Kimmel SE: Effect of antidepressants and their relative affinity for the serotonin transporter on the risk of myocardial infarction. Circulation 2003; 108:32–36CrossrefGoogle Scholar

157 Schultz SK, Anderson EA, van de Borne P: Heart rate variability before and after treatment with electroconvulsive therapy. J Affective Disord 1997; 44:13–20CrossrefGoogle Scholar

158 Nahshoni E, Aizenberg D, Sigler M, Zalsman G, Strasberg B, Imbar S, Weizman A: Heart rate variability in elderly patients before and after electroconvulsive therapy. Am J Geriatr Psychiatry 2001; 9:255–260CrossrefGoogle Scholar

159 Nahshoni E, Aizenberg D, Sigler M, Strasberg B, Zalsman G, Imbar S, Adler E, Weizman A: Heart rate variability increases in elderly depressed patients who respond to electroconvulsive therapy. J Psychosom Res 2004; 56:89–94CrossrefGoogle Scholar

160 Friedman M, Thoresen CE, Gill JJ, Ulsmer D, Powell LH, Price VA, Brown B, Thompson L, Rabin DD, Breall WS: Alteration of type A behavior and its effect on cardiac recurrences in post myocardial infarction patients: summary results of the Recurrent Coronary Prevention Project. Am Heart J 1986; 112:653–665CrossrefGoogle Scholar

161 Frasure-Smith N, Lesperance F, Prince RH, Verrier P, Garber RA, Juneau M, Wolfson C, Bourassa MA: Randomized trial of home-based psychosocial nursing intervention for patients recovering from myocardial infarction. Lancet 1997; 350:473–479CrossrefGoogle Scholar

162 Jones DA, West RR: Psychological rehabilitation after myocardial infarction: multicenter randomized controlled trial. BMJ 1996; 313:1517–1521CrossrefGoogle Scholar

163 Linden W, Stossel C, Maurice J: Psychological interventions for patients with coronary artery disease: a meta-analysis. Arch Intern Med 1996; 156:745–752CrossrefGoogle Scholar

164 Dusseldorp E, van Elderen T, Maes S, Meulman J, Kraaij V: A meta-analysis of psychoeduational programs for coronary heart disease patients. Health Psychology 1999; 18:506–519CrossrefGoogle Scholar

165 Schwartz PJ, Wolf S: QT interval prolongation as predictor of sudden death in patients with myocardial infarction. Circulation 1978; 57:1074–1077CrossrefGoogle Scholar

166 Algra A, Tijssen JG, Roelandt JR, Pool J, Lubsen J: QTc prolongation measured by standard 12-lead electrocardiography is an independent risk factor for sudden death due to cardiac arrest. Circulation 1991; 83:1888–1894CrossrefGoogle Scholar

167 Cardiac Arrhythmia Suppression Trial (CAST) Investigators: Preliminary report: effect of encainide and flecainide on mortality in a randomized trial of arrhythmia suppression after myocardial infarction. N Engl J Med 1989; 321:406–412CrossrefGoogle Scholar

168 Cardiac Arrhythmia Suppression Trial II Investigators: Effect of the antiarrhythmic agent moricizine on survival after myocardial infarction. N Engl J Med 1992; 327:227–233CrossrefGoogle Scholar

169 Muskin PR, Glassman AH: The use of tricyclic antidepressants in the medical setting, in Consultation-Liaison Psychiatry: Current Trends and Future Perspectives. Edited by Finkel JB. New York, Grune & Stratton, 1983, pp 137–158Google Scholar

170 Roose SP, Glassman AH, Giardina EGV, Walsh BT, Woodring S, Bigger JT: Tricyclic antidepressants in depressed patients with cardiac conduction disease. Arch Gen Psychiatry 1987; 44:273–275CrossrefGoogle Scholar

171 Roose SP, Glassman AH, Giardina EG, Johnson LL, Walsh BT, Bigger JT Jr: Cardiovascular effects of imipramine and bupropion in depressed patients with congestive heart failure. J Clin Psychopharmacol 1987; 7:247–251CrossrefGoogle Scholar

172 Glassman AH, Roose SP, Bigger JT Jr: The safety of tricyclic antidepressants in cardiac patients: risk-benefit reconsidered. JAMA 1993; 269:2673–2675CrossrefGoogle Scholar

173 Halperin P: Heart disease, in Textbook of Consultation-Liaison Psychiatry: Psychiatry in the Medically Ill, 2nd ed. Edited by Wise MG, Rundell JR. Washington, DC, American Psychiatric Publishing, 2002, pp 536–545Google Scholar

174 Rasmussen SL, Overo KF, Tanghoj P: Cardiac safety of citalopram: prospective trials and retrospective analyses. J Clin Psychopharmacol 1999; 19:407–415CrossrefGoogle Scholar

175 Hochberg HM, Kanter D, Houser VP: Electrocardiographic findings during extended clinical trials of fluvoxamine in depression: one years experience. Pharmacopsychiatry 1995; 28:253–256CrossrefGoogle Scholar

176 Baker B, Dorian P, Sandor P, Shapiro C, Schell C, Mitchell J, Irvine MJ: Electrocardiographic effects of fluoxetine and doxepin in patients with major depressive disorder. J Clin Psychopharmacol 1997; 17:15–21CrossrefGoogle Scholar

177 Roose SP, Laghrissi-Thode F, Kennedy JS, Nelson JC, Bigger JT, Jr, Pollock BG, Gaffney A, Narayan M, Finkel MS, McCafferty J, Gergel I: Comparison of paroxetine and nortriptyline in depressed patients with ischemic heart disease. JAMA 1998; 279:287–291CrossrefGoogle Scholar

178 Pacher P, Ungvari Z, Kecskemeti V, Furst S: Review of cardiovascular effects of fluoxetine, a selective serotonin reuptake inhibitor, compared to tricyclic antidepressants. Curr Med Chem 1998; 5:381–390Google Scholar

179 Graudins A, Vossler C, Wang R: Fluoxetine-induced cardiotoxicity with response to bicarbonate therapy. Am J Emerg Med 1997; 15:501–503CrossrefGoogle Scholar

180 Roose SP: Treatment of depression in patients with heart disease. Biol Psychiatry 2003; 54:262–268CrossrefGoogle Scholar

181 Davies SJC, Jackson PR, Potokar J, Nutt DJ: Treatment of anxiety and depressive disorders in patients with cardiovascular disease. BMJ 2004; 328:939–943CrossrefGoogle Scholar

182 Roose SP, Glassman AH, Attia E, Woodring S, Giardina E-GV, Bigger JT Jr: Cardiovascular effects of fluoxetine in depressed patients with heart disease. Am J Psychiatry 1998; 155:660–665CrossrefGoogle Scholar

183 Catalano G, Catalano MC, Epstein MA, Tsambiras PE: QTc interval prolongation associated with citalopram overdose: a case report and literature review. Clinical Neuropharmacol 2001; 24:158–162CrossrefGoogle Scholar

184 Isbister GK, Bowe SJ, Dawson A, Whyte IM: Relative toxicity of selective serotonin reuptake inhibitors (SSRIs) in overdose. J Toxicol Clin Toxicol 2004; 42:277–285CrossrefGoogle Scholar

185 Pacher P, Ungvari Z, Nanasi PP, Furst S, Kecskemeti V: Speculations on difference between tricyclic and selective serotonin reuptake inhibitor antidepressants on their cardiac effects: is there any? Curr Med Chem 1999; 6:469–480Google Scholar

186 Roose SP, Dalack GW, Glassman AH, Woodring S, Walsh BT, Giardina EGV: Cardiovascular effects of bupropion in depressed patients with heart disease. Am J Psychiatry 1991; 148:512–516CrossrefGoogle Scholar

187 Thase ME: Effects of venlafaxine on blood pressure: a meta-analysis of original data from 3744 depressed patients. J Clin Psychiatry 1998; 59:502–508CrossrefGoogle Scholar

188 Tulen JH, Bruijn JA, de Man KJ, Pepplinkhuizen L, van den Meiracker AH, Man in ’t Veld AJ: Cardiovascular variability in major depressive disorder and effects of imipramine or mirtazapine (Org 3770). J Clin Psychopharmacol 1996; 16:135–145CrossrefGoogle Scholar

189 Lesperance F, Frasure-Smith N, Laliberte MA, White M, Lafontaine S, Calderone A, Talajic M, Rouleau JL: An open-label study of nefazodone treatment of major depression in patients with congestive heart failure. Can J Psychiatry 2003; 40:695–701Google Scholar

190 Glassman AH, Bigger JT Jr: Antipsychotic drugs: prolonged QTc interval, torsade de pointes, and sudden death. Am J Psychiatry 2001; 158:1774–1782CrossrefGoogle Scholar

191 Reilly JG, Ayis SA, Ferrier IN, Jones SJ, Thomas SHL: Thioridazine and sudden unexplained death in psychiatric in-patients. Br J Psychiatry 2002; 180:515–522CrossrefGoogle Scholar

192 Piepho RW: Cardiovascular effects of antipsychotics used in bipolar illness. J Clin Psychiatry 2002; 63(suppl 4):20–23Google Scholar

193 Taylor DM: Antipsychotics and QT prolongation. Acta Psychiatr Scand 2003; 107:85–95CrossrefGoogle Scholar

194 Dewan NA, Suresh DP, Blomkalns A: Selecting safe psychotropics for post-MI patients. Curr Psychiatry 2003; 2:14–21Google Scholar

195 Fayek M, Kingsbury SJ, Zada J, Simpson GM: Psychopharmacology: cardiac effects of antipsychotic medications. Psychiatr Serv 2001; 52:607–609CrossrefGoogle Scholar

196 Chong S-A, Mythily. Arrhythmia and antipsychotic medications (letter). Psychiatr Serv 2001; 52:1257–1258CrossrefGoogle Scholar

197 Reilly JG, Ayis SA, Ferrier IN, Jones SJ, Thomas SHL: QTc-interval abnormalities and psychotropic drug therapy in psychiatric patients. Lancet 2000; 355(9209):1048–1052CrossrefGoogle Scholar

198 Vieweg WV: New generation antipsychotic drugs and QTc interval prolongation. Prim Care Companion J Clin Psychiatry 2003; 5:205–215CrossrefGoogle Scholar

199 Ravin DS, Levenson JW: Fatal cardiac event following initiation of risperidone therapy. Ann Pharmacother 1997; 31:867–870CrossrefGoogle Scholar

200 Al-Khatib SM, LaPointe NMA, Kramer JM, Califf RM: What clinicians should know about the QT interval. JAMA 2003; 289:2120–2127CrossrefGoogle Scholar

201 Moss AJ: Long QT syndrome. JAMA 2003; 289:2041–2044CrossrefGoogle Scholar

202 Vogel LR, Muskin PR, Collins ED, Sloan RP: Lorazepam reduces cardiac vagal modulation in normal subjects. J Clin Psychopharmacol 1996; 16:449–453CrossrefGoogle Scholar

203 Agelink MW, Majewski TB, Andrich J, Mueck-Weymann M: Short-term effects of intravenous benzodiazepines on autonomic neurocardiac regulation in humans: a comparison between midazolam, diazepam, and lorazepam. Crit Care Med 2002; 30:997–1006CrossrefGoogle Scholar

204 Unrug A, Bener J, Barry RJ, van Luijtelaar EL, Coenen AM, Kaiser J: Influence of diazepam and buspirone on human heart rate and the evoked cardiac response under varying cognitive load. Int J Psychophysiol 1997; 25:177–184CrossrefGoogle Scholar

205 Masand PS, Tesar GE: Use of stimulants in the medically ill. Psychiatr Clin North Am 1996; 19:515–548CrossrefGoogle Scholar

206 Masand PS, Murray GB, Pickett P: Psychostimulants in post-stroke depression. J Neuropsychiatry Clin Neurosci 1991; 3:23–27CrossrefGoogle Scholar

207 Mitchell JE, Mackenzie TB: Cardiac effects of lithium therapy in man: a review. J Clin Psychiatry 1982; 43:47–51Google Scholar

208 DasGupta K, Jefferson JW: The use of lithium in the medically ill. Gen Hosp Psychiatry 1990; 12:83–97CrossrefGoogle Scholar

209 McLaren K, Marangell L: Special considerations in the treatment of patients with bipolar disorder and medical comorbidities. Ann Gen Hosp Psychiatry 2004; 3:7CrossrefGoogle Scholar

210 Flockhart DA: Drug Interactions: Cytochrome P450 System. Available at http://www.drug-interactions.comGoogle Scholar

211 Shapiro PA: Heart disease, in: The American Psychiatric Publishing Textbook of Psychosom Medicine, 3rd ed. Edited by Levenson JL. Washington, DC, American Psychiatric Publishing, 2004, pp 423–444Google Scholar