Basic and Clinical Pharmacology, 13th Ed.

Antidepressant Agents

Charles DeBattista, MD

CASE STUDY

A 47-year-old woman presents to her primary care physician with a chief complaint of fatigue. She indicates that she was promoted to senior manager in her company approximately 11 months earlier. Although her promotion was welcome and came with a sizable raise in pay, it resulted in her having to move away from an office and group of colleagues she very much enjoyed. In addition, her level of responsibility increased dramatically. The patient reports that for the last 7 weeks, she has been waking up at 3 AM every night and been unable to go back to sleep. She dreads the day and the stresses of the workplace. As a consequence, she is not eating as well as she might and has dropped 7% of her body weight in the last 3 months. She also reports being so stressed that she breaks down crying in the office occasionally and has been calling in sick frequently. When she comes home, she finds she is less motivated to attend to chores around the house and has no motivation, interest, or energy to pursue recreational activities that she once enjoyed such as hiking. She describes herself as “chronically miserable and worried all the time.” Her medical history is notable for chronic neck pain from a motor vehicle accident for which she is being treated with tramadol and meperidine. In addition, she is on hydrochlorothiazide and propranolol for hypertension. The patient has a history of one depressive episode after a divorce that was treated successfully with fluoxetine. Medical workup including complete blood cell count, thyroid function tests, and a chemistry panel reveals no abnormalities. She is started on fluoxetine for a presumed major depressive episode and referred for cognitive behavioral psychotherapy. What CYP450 and pharmacodynamic interactions might be associated with fluoxetine use in this patient? Which class of antidepressants would be contraindicated in this patient?

The diagnosis of depression still rests primarily on the clinical interview. Major depressive disorder (MDD) is characterized by depressed mood most of the time for at least 2 weeks or loss of interest or pleasure in most activities, or both. In addition, depression is characterized by disturbances in sleep and appetite as well as deficits in cognition and energy. Thoughts of guilt, worthlessness, and suicide are common. Coronary artery disease, diabetes, and stroke appear to be more common in depressed patients, and depression may considerably worsen the prognosis for patients with a variety of comorbid medical conditions.

According to the Centers for Disease Control, antidepressants are consistently among the three most commonly prescribed classes of medications in the USA. The wisdom of such widespread use of antidepressants is debated. However, it is clear that American physicians have been increasingly inclined to use antidepressants to treat a host of conditions and that patients have been increasingly receptive to their use.

The primary indication for antidepressant agents is the treatment of MDD. Major depression, with a lifetime prevalence of around 17% in the USA and a point prevalence of 5%, is associated with substantial morbidity and mortality. MDD represents one of the most common causes of disability in the developed world. In addition, major depression is commonly associated with a variety of medical conditions—from chronic pain to coronary artery disease. When depression coexists with other medical conditions, the patient’s disease burden increases, and the quality of life—and often the prognosis for effective treatment—decreases significantly.

Some of the growth in antidepressant use may be related to the broad application of these agents for conditions other than major depression. For example, antidepressants have received FDA approvals for the treatment of panic disorder, generalized anxiety disorder (GAD), post-traumatic stress disorder (PTSD), and obsessive-compulsive disorder (OCD). In addition, antidepressants are commonly used to treat pain disorders such as neuropathic pain and the pain associated with fibromyalgia. Some antidepressants are used for treating premenstrual dysphoric disorder (PMDD), mitigating the vasomotor symptoms of menopause, and treating stress urinary incontinence. Thus, antidepressants have a broad spectrum of use in medical practice. However, their primary use remains the treatment for MDD.

Pathophysiology of Major Depression

There has been a marked shift in the last decade in our understanding of the pathophysiology of major depression. In addition to the older idea that a deficit in function or amount of monoamines (the monoamine hypothesis) is central to the biology of depression, there is evidence that neurotrophic and endocrine factors play a major role (the neurotrophic hypothesis). Histologic studies, structural and functional brain imaging research, genetic findings, and steroid research all suggest a complex pathophysiology for MDD with important implications for drug treatment.

Neurotrophic Hypothesis

There is substantial evidence that nerve growth factors such as brain-derived neurotrophic factor (BDNF) are critical in the regulation of neural plasticity, resilience, and neurogenesis. The evidence suggests that depression is associated with the loss of neurotrophic support and that effective antidepressant therapies increase neurogenesis and synaptic connectivity in cortical areas such as the hippocampus. BDNF is thought to exert its influence on neuronal survival and growth effects by activating the tyrosine kinase receptor B in both neurons and glia (Figure 30–1).

image

FIGURE 30–1 The neurotrophic hypothesis of major depression. Changes in trophic factors (especially brain-derived neurotrophic factor, BDNF) and hormones appear to play a major role in the development of major depression (A). Successful treatment results in changes in these factors (B). CREB, cAMP response element-binding (protein). BDNF, brain-derived neurotrophic factor. (Reproduced, with permission, from Nestler EJ: Neurobiology of depression. Neuron 2002;34[1]:13–25. Copyright Elsevier.)

Several lines of evidence support the neurotrophic hypothesis. Animal and human studies indicate that stress and pain are associated with a drop in BDNF levels and that this loss of neurotrophic support contributes to atrophic structural changes in the hippocampus and perhaps other areas such as the medial frontal cortex and anterior cingulate. The hippocampus is known to be important both in contextual memory and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Likewise, the anterior cingulate plays a role in the integration of emotional stimuli and attention functions, whereas the medial orbital frontal cortex is also thought to play a role in memory, learning, and emotion.

Over 30 structural imaging studies suggest that major depression is associated with a 5–10% loss of volume in the hippocampus, although some studies have not replicated this finding. Depression and chronic stress states have also been associated with a substantial loss of volume in the anterior cingulate and medial orbital frontal cortex. Loss of volume in structures such as the hippocampus also appears to increase as a function of the duration of illness and the amount of time that the depression remains untreated.

Another source of evidence supporting the neurotrophic hypothesis of depression comes from studies of the direct effects of BDNF on emotional regulation. Direct infusion of BDNF into the midbrain, hippocampus, and lateral ventricles of rodents has an antidepressant-like effect in animal models. Moreover, all known classes of antidepressants are associated with an increase in BDNF levels in animal models with chronic (but not acute) administration. This increase in BDNF levels is consistently associated with increased neurogenesis in the hippocampus in these animal models. Other interventions thought to be effective in the treatment of major depression, including electroconvulsive therapy, also appear to robustly stimulate BDNF levels and hippocampus neurogenesis in animal models.

Human studies seem to support the animal data on the role of neurotrophic factors in stress states. Depression appears to be associated with a drop in BDNF levels in the cerebrospinal fluid and serum as well as with a decrease in tyrosine kinase receptor B activity. Conversely, administration of antidepressants increases BDNF levels in clinical trials and may be associated with an increase in hippocampus volume in some patients.

Much evidence supports the neurotrophic hypothesis of depression, but not all evidence is consistent with this concept. Animal studies in BDNF knockout mice have not always suggested an increase in depressive or anxious behaviors that would be expected with a deficiency of BDNF. In addition, some animal studies have found an increase in BDNF levels after some types of social stress and an increase rather than a decrease in depressive behaviors with lateral ventricle injections of BDNF.

A proposed explanation for the discrepant findings on the role of neurotrophic factors in depression is that there are polymorphisms for BDNF that may yield very different effects. Mutations in the BDNFgene have been found to be associated with altered anxiety and depressive behavior in both animal and human studies.

Thus, the neurotrophic hypothesis continues to be intensely investigated and has yielded new insights and potential targets in the treatment of MDD.

Monoamines & Other Neurotransmitters

The monoamine hypothesis of depression (Figure 30–2) suggests that depression is related to a deficiency in the amount or function of cortical and limbic serotonin (5-HT), norepinephrine (NE), and dopamine (DA).

image

FIGURE 30–2 The amine hypothesis of major depression. Depression appears to be associated with changes in serotonin or norepinephrine signaling in the brain (or both) with significant downstream effects. Most antidepressants cause changes in amine signaling. AC, adenylyl cyclase; 5-HT, serotonin; CREB, cAMP response element-binding (protein); DAG, diacyl glycerol; IP3, inositol trisphosphate; MAO, monoamine oxidase; NET, norepinephrine transporter; PKC, protein kinase C; PLC, phospholipase C; SERT, serotonin transporter. (Adapted from Belmaker R, Agam G: Major depressive disorder. N Engl J Med 2008;358:59.)

Evidence to support the monoamine hypothesis comes from several sources. It has been known for many years that reserpine treatment, which is known to deplete monoamines, is associated with depression in a subset of patients. Similarly, depressed patients who respond to serotonergic antidepressants such as fluoxetine often rapidly suffer relapse when given diets free of tryptophan, a precursor of serotonin synthesis. Patients who respond to noradrenergic antidepressants such as desipramine are less likely to relapse on a tryptophan-free diet. Moreover, depleting catecholamines in depressed patients who have previously responded to noradrenergic agents likewise tends to be associated with relapse. Administration of an inhibitor of norepinephrine synthesis is also associated with a rapid return of depressive symptoms in patients who respond to noradrenergic but not necessarily in patients who had responded to serotonergic antidepressants.

Another line of evidence supporting the monoamine hypothesis comes from genetic studies. A functional polymorphism exists for the promoter region of the serotonin transporter gene, which regulates how much of the transporter protein is available. Subjects who are homozygous for the s (short) allele may be more vulnerable to developing major depression and suicidal behavior in response to stress. In addition, homozygotes for the s allele may also be less likely to respond to and tolerate serotonergic antidepressants. Conversely, subjects with the l (long) allele tend to be more resistant to stress and may be more likely to respond to serotonergic antidepressants.

Studies of depressed patients have sometimes shown an alteration in monoamine function. For example, some studies have found evidence of alteration in serotonin receptor numbers (5-HT1A and 5-HT2C) or norepinephrine (α2) receptors in depressed and suicidal patients, but these findings have not been consistent. A reduction in the primary serotonin metabolite 5-hydroxyindoleacetic acid in the cerebrospinal fluid is associated with violent and impulsive behavior, including violent suicide attempts. However, this finding is not specific to major depression and is associated more generally with violent and impulsive behavior.

Finally, perhaps the most convincing line of evidence supporting the monoamine hypothesis is the fact that (at the time of this writing) all available antidepressants appear to have significant effects on the monoamine system. All classes of antidepressants appear to enhance the synaptic availability of 5-HT, norepinephrine, or dopamine. Attempts to develop antidepressants that work on other neurotransmitter systems have not been effective to date.

The monoamine hypothesis, like the neurotrophic hypothesis, is at best incomplete. Many studies have not found an alteration in function or levels of monoamines in depressed patients. In addition, some candidate antidepressant agents under study do not act directly on the monoamine system.

In addition to the monoamines, the excitatory neurotransmitter glutamate appears to be important in the pathophysiology of depression. A number of studies of depressed patients have found elevated glutamate content in the cerebrospinal fluid of depressed patients and decreased glutamine/glutamate ratios in their plasma. In addition, postmortem studies have revealed significant increases in the frontal and dorsolateral prefrontal cortex of depressed patients. Likewise, structural neuroimaging studies have consistently found volumetric changes in the brain areas of depressed patients in which glutamate neurons and their connections are most abundant, including the amygdala and hippocampus.

Antidepressants are known to impact glutamate neurotransmission in a variety of ways. For example, chronic antidepressant use is associated with reducing glutamatergic transmission, including the presynaptic release of glutamate in the hippocampus and cortical areas. Similarly, the chronic administration of antidepressants significantly reduces depolarization-evoked release of glutamate in animal models. Stress is known to enhance the release of glutamate in rodents, and antidepressants inhibit stress-induced presynaptic release of glutamate in these models.

Given the effect of antidepressants on the glutamate system, there has been a growing interest in the development of pharmaceutical agents that might modulate the glutamate system. Ketamine is a potent, high-affinity, noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that has long been used in anesthesia and is a common drug of abuse in some parts of the world. A number of preclinical and clinical studies have demonstrated rapid antidepressant effects of ketamine. Multiple studies have suggested that a single dose of intravenous ketamine at subanesthetic doses produces rapid relief of depression, even in treatment-resistant patients, that may persist for 1 week or longer. Unfortunately, ketamine is associated with cognitive, dissociative, and psychotomimetic properties that make it impractical as a long-term treatment for depression. Still, a number of other NMDA receptor antagonists, partial antagonists, and metabotropic glutamate receptor modulators (see Chapter 29) are under investigation as potential antidepressants.

Neuroendocrine Factors in the Pathophysiology of Depression

Depression is known to be associated with a number of hormonal abnormalities. Among the most replicated of these findings are abnormalities in the HPA axis in patients with MDD. Moreover, MDD is associated with elevated cortisol levels (Figure 30–1), nonsuppression of adrenocorticotropic hormone (ACTH) release in the dexamethasone suppression test, and chronically elevated levels of corticotropin-releasing hormone. The significance of these HPA abnormalities is unclear, but they are thought to indicate a dysregulation of the stress hormone axis. More severe types of depression, such as psychotic depression, tend to be associated with HPA abnormalities more commonly than milder forms of major depression. It is well known that both exogenous glucocorticoids and endogenous elevation of cortisol are associated with mood symptoms and cognitive deficits similar to those seen in MDD.

Thyroid dysregulation has also been reported in depressed patients. Up to 25% of depressed patients are reported to have abnormal thyroid function. These abnormalities include a blunting of response of thyrotropin to thyrotropin-releasing hormone, and elevations in circulating thyroxine during depressed states. Clinical hypothyroidism often presents with depressive symptoms, which resolve with thyroid hormone supplementation. Thyroid hormones are also commonly used in conjunction with standard antidepressants to augment therapeutic effects of the latter.

Finally, sex steroids are also implicated in the pathophysiology of depression. Estrogen deficiency states, which occur in the postpartum and postmenopausal periods, are thought to play a role in the etiology of depression in some women. Likewise, severe testosterone deficiency in men is sometimes associated with depressive symptoms. Hormone replacement therapy in hypogonadal men and women may be associated with an improvement in mood and depressive symptoms.

Integration of Hypotheses Regarding the Pathophysiology of Depression

The several pathophysiologic hypotheses just described are not mutually exclusive. It is evident that the monoamine, neuroendocrine, and neurotrophic systems are interrelated in important ways. For example, HPA and steroid abnormalities may contribute to suppression of transcription of the BDNF gene. Glucocorticoid receptors are found in high density in the hippocampus. Binding of these hippocampal glucocorticoid receptors by cortisol during chronic stress states such as major depression may decrease BDNF synthesis and may result in volume loss in stress-sensitive regions such as the hippocampus. The chronic activation of monoamine receptors by antidepressants appears to have the opposite effect of stress and results in an increase in BDNF transcription. In addition, activation of monoamine receptors appears to down-regulate the HPA axis and may normalize HPA function.

One of the weaknesses of the monoamine hypothesis is the fact that amine levels increase immediately with antidepressant use, but maximum beneficial effects of most antidepressants are not seen for many weeks. The time required to synthesize neurotrophic factors has been proposed as an explanation for this delay of antidepressant effects. Appreciable protein synthesis of products such as BDNF typically takes 2 weeks or longer and coincides with the clinical course of antidepressant treatment.

image BASIC PHARMACOLOGY OF ANTIDEPRESSANTS

Chemistry & Subgroups

The currently available antidepressants make up a remarkable variety of chemical types. These differences and the differences in their molecular targets provide the basis for distinguishing several subgroups.

A. Selective Serotonin Reuptake Inhibitors

The selective serotonin reuptake inhibitors (SSRIs) represent a chemically diverse class of agents that have as their primary action the inhibition of the serotonin transporter (SERT; Figure 30–3). Fluoxetine was introduced in the United States in 1988 and quickly became one of the most commonly prescribed medications in medical practice. The development of fluoxetine emerged out of the search for chemicals that had high affinity for monoamine receptors but lacked the affinity for histamine, acetylcholine, and α adrenoceptors that is seen with the tricyclic antidepressants (TCAs). There are currently six available SSRIs, and they are the most common antidepressants in clinical use. In addition to their use in major depression, SSRIs have indications in GAD, PTSD, OCD, panic disorder, PMDD, and bulimia. Fluoxetine, sertraline, and citalopram exist as isomers and are formulated in the racemic forms, whereas paroxetine and fluvoxamine are not optically active. Escitalopram is the (S) enantiomer of citalopram. As with all antidepressants, SSRIs are highly lipophilic. The popularity of SSRIs stems largely from their ease of use, safety in overdose, relative tolerability, cost (all are available as generic products), and broad spectrum of uses.

image

FIGURE 30–3 Structures of several selective serotonin reuptake inhibitors (SSRIs).

B. Serotonin-Norepinephrine Reuptake Inhibitors

Two classes of antidepressants act as combined serotonin and norepinephrine reuptake inhibitors: selective serotonin-norepinephrine reuptake inhibitors (SNRIs) and TCAs.

1. Selective serotonin-norepinephrine reuptake inhibitorsThe SNRIs include venlafaxine, its metabolite desvenlafaxine, duloxetine, and levomilnacipran. Levomilnacipran is the active enantiomer of a racemic SNRI, milnacipran. Milnacipran has been approved for the treatment of fibromyalgia in the USA and has been used in the treatment of depression in Europe for many years. In addition to their use in major depression, SNRIs have applications in the treatment of pain disorders including neuropathies and fibromyalgia. SNRIs are also used in the treatment of generalized anxiety, stress urinary incontinence, and vasomotor symptoms of menopause.

image

SNRIs are chemically unrelated to each other. Venlafaxine was discovered in the process of evaluating chemicals that inhibit binding of imipramine. Venlafaxine’s in vivo effects are similar to those of imipramine but with a more favorable adverse-effect profile. All SNRIs bind the serotonin (SERT) and norepinephrine (NET) transporters, as do the TCAs. However, unlike the TCAs, the SNRIs do not have much affinity for other receptors. Venlafaxine and desvenlafaxine are bicyclic compounds, whereas duloxetine is a three-ring structure unrelated to the TCAs. Milnacipran contains a cyclopropane ring and is provided as a racemic mixture.

image

2. Tricyclic antidepressantsThe TCAs were the dominant class of antidepressants until the introduction of SSRIs in the 1980s and 1990s. Nine TCAs are available in the USA, and they all have an iminodibenzyl (tricyclic) core (Figure 30–4). The chemical differences between the TCAs are relatively subtle. For example, the prototype TCA imipramine and its metabolite, desipramine, differ by only a methyl group in the propylamine side chain. However, this minor difference results in a substantial change in their pharmacologic profiles. Imipramine is highly anticholinergic and is a relatively strong serotonin as well as norepinephrine reuptake inhibitor. In contrast, desipramine is much less anticholinergic and is a more potent and somewhat more selective norepinephrine reuptake inhibitor than is imipramine.

image

FIGURE 30–4 Structures of some tricyclic antidepressants (TCAs).

At the present time, the TCAs are used primarily in depression that is unresponsive to more commonly used antidepressants such as the SSRIs or SNRIs. Their loss of popularity stems in large part from relatively poorer tolerability compared with newer agents, difficulty of use, and lethality in overdose. Other uses for TCAs include the treatment of pain conditions, enuresis, and insomnia.

C. 5-HT2 Receptor Modulators

Two antidepressants are thought to act primarily as antagonists at the 5-HT2 receptor: trazodone and nefazodone. Trazodone’s structure includes a triazolo moiety that is thought to impart antidepressant effects. Its primary metabolite, m-chlorphenylpiperazine (m-cpp), is a potent 5-HT2 antagonist. Trazodone was among the most commonly prescribed antidepressants until it was supplanted by the SSRIs in the late 1980s. The most common use of trazodone in current practice is as an unlabeled hypnotic, since it is highly sedating and not associated with tolerance or dependence.

image

Nefazodone is chemically related to trazodone. Its primary metabolites, hydroxynefazodone and m-cpp are both inhibitors of the 5-HT2 receptor. Nefazodone received an FDA black box warning in 2001 implicating it in hepatotoxicity, including lethal cases of hepatic failure. Though still available generically, nefazodone is no longer commonly prescribed. The primary indications for both nefazodone and trazodone are major depression, although both have also been used in the treatment of anxiety disorders.

image

Vortioxetine is a newer agent that acts as an antagonist of the 5-HT3, 5-HT7, and 5-HT1D receptors, a partial agonist of the 5-HT1B receptor, and an agonist of the 5HT1A receptor. It also inhibits the serotonin transporter but its actions are not primarily related to SERT inhibition and it is therefore not classified as an SSRI. Vortioxetine has demonstrated efficacy on major depression in a number of controlled clinical studies. In addition, there is some preliminary evidence that the drug also may improve some aspects of cognition in depressed patients.

D. Tetracyclic and Unicyclic Antidepressants

A number of antidepressants do not fit neatly into the other classes. Among these are bupropion, mirtazapine, amoxapine, vilazodone, and maprotiline (Figure 30–5). Bupropion has a unicyclic aminoketone structure. Its unique structure results in a different side-effect profile than most antidepressants (described below). Bupropion somewhat resembles amphetamine in chemical structure and, like the stimulant, has central nervous system (CNS) activating properties.

image

FIGURE 30–5 Structures of the tetracyclics, amoxapine, maprotiline, and mirtazapine and the unicyclic, bupropion.

Mirtazapine was introduced in 1994 and, like bupropion, is one of the few antidepressants not commonly associated with sexual effects. It has a tetracyclic chemical structure and belongs to the piperazino-azepine group of compounds.

Mirtazapine, amoxapine, and maprotiline have tetracyclic structures. Amoxapine is the N-methylated metabolite of loxapine, an older antipsychotic drug. Amoxapine and maprotiline share structural similarities and side effects comparable to the TCAs. As a result, these tetracyclics are not commonly prescribed in current practice. Their primary use is in MDD that is unresponsive to other agents. Vilazodone has a multi-ring structure that allows it to bind potently to the serotonin transporter but minimally to the dopamine and norepinephrine transporter.

E. Monoamine Oxidase Inhibitors

Arguably the first modern class of antidepressants, monoamine oxidase inhibitors (MAOIs) were introduced in the 1950s but are now rarely used in clinical practice because of toxicity and potentially lethal food and drug interactions. Their primary use now is in the treatment of depression unresponsive to other antidepressants. However, MAOIs have also been used historically to treat anxiety states, including social anxiety and panic disorder. In addition, selegiline is used in the treatment of Parkinson’s disease (see Chapter 28).

Current MAOIs include the hydrazine derivatives phenelzine and isocarboxazid and the non-hydrazines tranylcypromine, selegiline, and moclobemide (the latter is not available in the USA). The hydrazines and tranylcypromine bind irreversibly and nonselectively with MAO-A and -B, whereas other MAOIs may have more selective or reversible properties. Some of the MAOIs such as tranylcypromine resemble amphetamine in chemical structure, whereas other MAOIs such as selegiline have amphetamine-like metabolites. As a result, these MAOIs tend to have substantial CNS-stimulating effects.

image

Pharmacokinetics

The antidepressants share several pharmacokinetic features (Table 30–1). Most have fairly rapid oral absorption, achieve peak plasma levels within 2–3 hours, are tightly bound to plasma proteins, undergo hepatic metabolism, and are renally cleared. However, even within classes, the pharmacokinetics of individual antidepressants varies considerably.

TABLE 30–1 Pharmacokinetic profiles of selected antidepressants.

image

A. Selective Serotonin Reuptake Inhibitors

The prototype SSRI, fluoxetine, differs from other SSRIs in some important respects (Table 30–1). Fluoxetine is metabolized to an active product, norfluoxetine, which may have plasma concentrations greater than those of fluoxetine. The elimination half-life of norfluoxetine is about three times longer than fluoxetine and contributes to the longest half-life of all the SSRIs. As a result, fluoxetine has to be discontinued 4 weeks or longer before an MAOI can be administered to mitigate the risk of serotonin syndrome.

Fluoxetine and paroxetine are potent inhibitors of the CYP2D6 isoenzyme, and this contributes to potential drug interactions (see Drug Interactions). In contrast, fluvoxamine is an inhibitor of CYP3A4, whereas citalopram, escitalopram, and sertraline have more modest CYP interactions.

B. Serotonin-Norepinephrine Reuptake Inhibitors

1. Selective serotonin-norepinephrine reuptake inhibitorsVenlafaxine is extensively metabolized in the liver via the CYP2D6 isoenzyme to O-desmethylvenlafaxine (desvenlafaxine). Both have similar half-lives of about 8-11 hours. Despite the relatively short half-lives, both drugs are available in formulations that allow once-daily dosing. Venlafaxine and desvenlafaxine have the lowest protein binding of all antidepressants (27–30%). Unlike most antidepressants, desvenlafaxine is conjugated and does not undergo extensive oxidative metabolism. At least 45% of desvenlafaxine is excreted unchanged in the urine compared with 4–8% of venlafaxine.

Duloxetine is well absorbed and has a half-life of 12-15 hours but is dosed once daily. It is tightly bound to protein (97%) and undergoes extensive oxidative metabolism via CYP2D6 and CYP1A2. Hepatic impairment significantly alters duloxetine levels unlike desvenlafaxine.

Both milnacipran and levomilnacipran are well absorbed after oral dosing.Both have shorter half-lives and lower protein binding than venlafaxine (Table 30–1). Milnacipran and levomilnacipran are largely excreted unchanged in the urine. Levomilnacipran also undergoes desethylation via 3A3/4.

2. Tricyclic antidepressantsThe TCAs tend to be well absorbed and have long half-lives (Table 30–1). As a result, most are dosed once daily at night because of their sedating effects. TCAs undergo extensive metabolism via demethylation, aromatic hydroxylation, and glucuronide conjugation. Only about 5% of TCAs are excreted unchanged in the urine. The TCAs are substrates of the CYP2D6 system, and the serum levels of these agents tend to be substantially influenced by concurrent administration of drugs such as fluoxetine. In addition, genetic polymorphism for CYP2D6 may result in low or extensive metabolism of the TCAs.

The secondary amine TCAs, including desipramine and nortriptyline, lack active metabolites and have fairly linear kinetics. These TCAs have a wide therapeutic window, and serum levels are reliable in predicting response and toxicity.

C. 5-HT Receptor Modulators

Trazodone and nefazodone are rapidly absorbed and undergo hepatic metabolism. Both drugs are bound to protein and have limited bioavailability because of extensive metabolism. Because of their short half-lives split dosing is generally required when these drugs are used as antidepressants. However, trazodone is often prescribed as a single dose at night as a hypnotic in lower doses than are used in the treatment of depression. Both trazodone and nefazodone have active metabolites that also exhibit 5-HT2 antagonism. Nefazodone is a potent inhibitor of the CYP3A4 system and may interact with drugs metabolized by this enzyme (see Drug Interactions). Vortioxetine is not a potent inhibitor of CYP isoenzymes. However, it is extensively metabolized through oxidation by CYP2D6 and other isoenzymes and then undergoes subsequent glucuronic acid conjugation. It is tightly bound to protein and has linear and dose-proportional pharmacokinetics.

D. Tetracyclic and Unicyclic Agents

Bupropion is rapidly absorbed and has a mean protein binding of 85%. It undergoes extensive hepatic metabolism and has a substantial first-pass effect. It has three active metabolites including hydroxybupropion; the latter is being developed as an antidepressant. Bupropion has a biphasic elimination with the first phase lasting about 1 hour and the second phase lasting 14 hours.

Amoxapine is also rapidly absorbed with protein binding of about 85%. The half-life is variable, and the drug is often given in divided doses. Amoxapine undergoes extensive hepatic metabolism. One of the active metabolites, 7-hydroxyamoxapine, is a potent D2 blocker and is associated with antipsychotic effects. Maprotiline is similarly well absorbed orally and 88% bound to protein. It undergoes extensive hepatic metabolism.

Mirtazapine is demethylated followed by hydroxylation and glucuronide conjugation. Several CYP isozymes are involved in the metabolism of mirtazapine, including 2D6, 3A4, and 1A2. The half-life of mirtazapine is 20–40 hours, and it is usually dosed once in the evening because of its sedating effects.

Vilazodone is well absorbed (Table 30–1) and absorption is increased when it is given with a fatty meal. It is extensively metabolized by CYP3A4 with minor contributions by CYP2C19 and CYP2D6. Only 1% of vilazodone is excreted unchanged in the urine.

E. Monoamine Oxidase Inhibitors

The different MAOIs are metabolized via different pathways but tend to have extensive first-pass effects that may substantially decrease bioavailability. Tranylcypromine is ring hydroxylated and N-acetylated, whereas acetylation appears to be a minor pathway for phenelzine. Selegiline is N-demethylated and then hydroxylated. The MAOIs are well absorbed from the gastrointestinal tract.

Because of the prominent first-pass effects and their tendency to inhibit MAO in the gut (resulting in tyramine pressor effects), alternative routes of administration are being developed. For example, selegiline is available in both transdermal and sublingual forms that bypass both gut and liver. These routes decrease the risk of food interactions and provide substantially increased bioavailability.

Pharmacodynamics

As previously noted, all currently available antidepressants enhance monoamine neurotransmission by one of several mechanisms. The most common mechanism is inhibition of the activity of SERT, NET, or both monoamine transporters (Table 30–2). Antidepressants that inhibit SERT, NET, or both include the SSRIs and SNRIs (by definition), and the TCAs. Another mechanism for increasing the availability of monoamines is inhibition of their enzymatic degradation (by the MAOIs). Additional strategies for enhancing monoamine tone include binding presynaptic autoreceptors (mirtazapine) or specific postsynaptic receptors (5-HT2 antagonists and mirtazapine). Ultimately, the increased availability of monoamines for binding in the synaptic cleft results in a cascade of events that enhance the transcription of some proteins and the inhibition of others. It is the net production of these proteins, including BDNF, glucocorticoid receptors, β adrenoceptors, and other proteins, that appears to determine the benefits as well as the toxicity of a given agent.

TABLE 30–2 Antidepressant effects on several receptors and transporters.

image

A. Selective Serotonin Reuptake Inhibitors

The serotonin transporter (SERT) is a glycoprotein with 12 transmembrane regions embedded in the axon terminal and cell body membranes of serotonergic neurons. When extracellular serotonin binds to receptors on the transporter, conformational changes occur in the transporter and serotonin, Na+, and Cl- are moved into the cell. Binding of intracellular K+ then results in the release of serotonin inside the cell and return of the transporter to its original conformation. SSRIs allosterically inhibit the transporter by binding the SERT receptor at a site other than the serotonin binding site. At therapeutic doses, about 80% of the activity of the transporter is inhibited. Functional polymorphisms exist for SERT that determine the activity of the transporter (Table 30–2).

SSRIs have modest effects on other neurotransmitters. Unlike TCAs and SNRIs, there is little evidence that SSRIs have prominent effects on β adrenoceptors or the norepinephrine transporter, NET. Binding to the serotonin transporter is associated with tonic inhibition of the dopamine system, although there is substantial interindividual variability in this effect. The SSRIs do not bind aggressively to histamine, muscarinic, or other receptors.

B. Drugs That Block Both Serotonin and Norepinephrine Transporters

A large number of antidepressants have mixed inhibitory effects on both serotonin and norepinephrine transporters. The newer agents in this class (venlafaxine and duloxetine) are termed SNRIs; those in the older group are termed TCAs on the basis of their structures.

1. Serotonin-norepinephrine reuptake inhibitorsSNRIs bind both the serotonin and the norepinephrine transporters. The NET is structurally very similar to the 5-HT transporter. Like the serotonin transporter, it is a 12-transmembrane domain complex that allosterically binds norepinephrine. The NET also has a moderate affinity for dopamine.

Venlafaxine is a weak inhibitor of NET, whereas desvenlafaxine, duloxetine, milnacipran, and levomilnacipran are more balanced inhibitors of both SERT and NET. Nonetheless, the affinity of most SNRIs tends to be much greater for SERT than for NET. The SNRIs differ from the TCAs in that they lack the potent antihistamine, α-adrenergic blocking, and anticholinergic effects of the TCAs. As a result, the SNRIs tend to be favored over the TCAs in the treatment of MDD and pain syndromes because of their better tolerability.

2. Tricyclic antidepressantsThe TCAs resemble the SNRIs in function, and their antidepressant activity is thought to relate primarily to their inhibition of 5-HT and norepinephrine reuptake. Within the TCAs, there is considerable variability in affinity for SERT versus NET. For example, clomipramine has relatively very little affinity for NET but potently binds SERT. This selectivity for the serotonin transporter contributes to clomipramine’s known benefits in the treatment of OCD. On the other hand, the secondary amine TCAs, desipramine and nortriptyline, are relatively more selective for NET. Although the tertiary amine TCA imipramine has more serotonin effect initially, its metabolite, desipramine, then balances this effect with more NET inhibition.

Common adverse effects of the TCAs, including dry mouth and constipation, are attributable to the potent antimuscarinic effects of many of these drugs. The TCAs also tend to be potent antagonists of the histamine H1 receptor. TCAs such as doxepin are sometimes prescribed as hypnotics and used in treatments for pruritus because of their antihistamine properties. The blockade of α adrenoceptors can result in substantial orthostatic hypotension, particularly in older patients.

C. 5-HT Receptor Modulators

The principle action of both nefazodone and trazodone appears to be blockade of the 5-HT2A receptor. Inhibition of this receptor in both animal and human studies is associated with substantial antianxiety, antipsychotic, and antidepressant effects. Conversely, agonists of the 5-HT2A receptor, eg, lysergic acid (LSD) and mescaline, are often hallucinogenic and anxiogenic. The 5-HT2A receptor is a G protein-coupled receptor and is distributed throughout the neocortex.

Nefazodone is a weak inhibitor of both SERT and NET but is a potent antagonist of the postsynaptic 5-HT2A receptor, as are its metabolites. Trazodone is also a weak but selective inhibitor of SERT with little effect on NET. Its primary metabolite, m-cpp, is a potent 5-HT2 antagonist, and much of trazodone’s benefits as an antidepressant might be attributed to this effect. Trazodone also has weak-to-moderate presynaptic α-adrenergic–blocking properties and is a modest antagonist of the H1 receptor.

As described above, vortioxetine has multimodal effects on a variety of 5-HT receptors and is an allosteric inhibitor of SERT. It has no known direct activity on norepinephrine or dopamine receptors.

D. Tetracyclic and Unicyclic Antidepressants

The actions of bupropion remain poorly understood. Bupropion and its major metabolite hydroxybupropion are modest-to-moderate inhibitors of norepinephrine and dopamine reuptake in animal studies. However, these effects seem less than are typically associated with antidepressant benefit. A more significant effect of bupropion is presynaptic release of catecholamines. In animal studies, bupropion appears to substantially increase the presynaptic availability of norepinephrine, and dopamine to a lesser extent. Bupropion has virtually no direct effects on the serotonin system.

Mirtazapine has a complex pharmacology. It is an antagonist of the presynaptic α2 autoreceptor and enhances the release of both norepinephrine and 5-HT. In addition, mirtazapine is an antagonist of 5-HT2and 5-HT3 receptors. Finally, mirtazapine is a potent H1 antagonist, which is associated with the drug’s sedative effects.

The actions of amoxapine and maprotiline resemble those of TCAs such as desipramine. Both are potent NET inhibitors and less potent SERT inhibitors. In addition, both possess anticholinergic properties. Unlike the TCAs or other antidepressants, amoxapine is a moderate inhibitor of the postsynaptic D2 receptor. As such, amoxapine possesses some antipsychotic properties.

Vilazodone is a potent serotonin reuptake inhibitor and a partial agonist of the 5-HT1A receptor. Partial agonists of the 5-HT1A receptor such as buspirone are thought to have mild to moderate antidepressant and anxiolytic properties.

E. Monoamine Oxidase Inhibitors

MAOIs act by mitigating the actions of monoamine oxidase in the neuron and increasing monoamine content. There are two forms of monoamine oxidase. MAO-A is present in both dopamine and norepinephrine neurons and is found primarily in the brain, gut, placenta, and liver; its primary substrates are norepinephrine, epinephrine, and serotonin. MAO-B is found primarily in serotonergic and histaminergic neurons and is distributed in the brain, liver, and platelets. MAO-B acts primarily on dopamine, tyramine, phenylethylamine, and benzylamine. Both MAO-A and -B metabolize tryptamine.

MAOIs are classified by their specificity for MAO-A or -B and whether their effects are reversible or irreversible. Phenelzine and tranylcypromine are examples of irreversible, nonselective MAOIs. Moclobemide is a reversible and selective inhibitor of MAO-A but is not available in the USA. Moclobemide can be displaced from MAO-A by tyramine, and this mitigates the risk of food interactions. In contrast, selegiline is an irreversible MAO-B–specific agent at low doses. Selegiline is useful in the treatment of Parkinson’s disease at these low doses, but at higher doses it becomes a nonselective MAOI similar to other agents.

image CLINICAL PHARMACOLOGY OF ANTIDEPRESSANTS

Clinical Indications

A. Depression

The FDA indication for the use of the antidepressants in the treatment of major depression is fairly broad. Most antidepressants are approved for both acute and long-term treatment of major depression. Acute episodes of MDD tend to last about 6–14 months untreated, but at least 20% of episodes last 2 years or longer.

The goal of acute treatment of MDD is remission of all symptoms. Since antidepressants may not achieve their maximum benefit for 1–2 months or longer, it is not unusual for a trial of therapy to last 8–12 weeks at therapeutic doses. The antidepressants are successful in achieving remission in about 30–40% of patients within a single trial of 8–12 weeks. If an inadequate response is obtained, therapy is often switched to another agent or augmented by addition of another drug. For example, bupropion, an atypical antipsychotic, or mirtazapine might be added to an SSRI or SNRI to augment antidepressant benefit if monotherapy is unsuccessful. Seventy to eighty percent of patients are able to achieve remission with sequenced augmentation or switching strategies. Once an adequate response is achieved, continuation therapy is recommended for a minimum of 6–12 months to reduce the substantial risk of relapse.

Approximately 85% of patients who have a single episode of MDD will have at least one recurrence in a lifetime. Many patients have multiple recurrences, and these recurrences may progress to more serious, chronic, and treatment-resistant episodes. Thus, it is not unusual for patients to require maintenance treatment to prevent recurrences. Although maintenance treatment studies of more than 5 years are uncommon, long-term studies with TCAs, SNRIs, and SSRIs suggest a significant protective benefit when given chronically. Thus, it is commonly recommended that patients be considered for long-term maintenance treatment if they have had two or more serious MDD episodes in the previous 5 years or three or more serious episodes in a lifetime.

It is not clear whether antidepressants are useful for all subtypes of depression. For example, patients with bipolar depression may not benefit much from antidepressants even when added to mood stabilizers. In fact, the antidepressants are sometimes associated with switches into mania or more rapid cycling. There has also been some debate about the overall efficacy of antidepressants in unipolar depression, with some meta-analyses showing large effects and others showing more modest effects. Although this debate is not likely to be settled immediately, there is little debate that antidepressants have important benefits for most patients.

Psychotherapeutic interventions such as cognitive behavioral therapy appear to be as effective as antidepressant treatment for mild to moderate forms of depression. However, cognitive behavioral therapy tends to take longer to be effective and is generally more expensive than antidepressant treatment. Psychotherapy is often combined with antidepressant treatment, and the combination appears more effective than either strategy alone.

B. Anxiety Disorders

After major depression, anxiety disorders represent the most common application of antidepressants. A number of SSRIs and SNRIs have been approved for all the major anxiety disorders, including PTSD, OCD, social anxiety disorder, GAD, and panic disorder. Panic disorder is characterized by recurrent episodes of brief overwhelming anxiety, which often occur without precipitant. Patients may begin to fear having an attack, or they avoid situations in which they might have an attack. In contrast, GAD is characterized by a chronic, free-floating anxiety and undue worry that tends to be chronic in nature. Although older antidepressants and drugs of the sedative-hypnotic class are still occasionally used for the treatment of anxiety disorders, SSRIs and SNRIs have largely replaced them.

The benzodiazepines (see Chapter 22) provide much more rapid relief of both generalized anxiety and panic than do any of the antidepressants. However, the antidepressants appear to be at least as effective as, and perhaps more effective than, benzodiazepines in the long-term treatment of these anxiety disorders. Furthermore, antidepressants do not carry the risks of dependence and tolerance that may occur with the benzodiazepines.

OCD is known to respond to serotonergic antidepressants. It is characterized by repetitive anxiety-provoking thoughts (obsessions) or repetitive behaviors aimed at reducing anxiety (compulsions). Clomipramine and several of the SSRIs are approved for the treatment of OCD, and they are moderately effective. Behavior therapy is usually combined with the antidepressant for additional benefits.

Social anxiety disorder is an uncommonly diagnosed but a fairly common condition in which patients experience severe anxiety in social interactions. This anxiety may limit their ability to function adequately in their jobs or interpersonal relationships. Several SSRIs and venlafaxine are approved for the treatment of social anxiety. The efficacy of the SSRIs in the treatment of social anxiety is greater in some studies than their efficacy in the treatment of MDD.

PTSD is manifested when a traumatic or life-threatening event results in intrusive anxiety-provoking thoughts or imagery, hypervigilance, nightmares, and avoidance of situations that remind the patient of the trauma. SSRIs are considered first-line treatment for PTSD and can benefit a number of symptoms including anxious thoughts and hypervigilance. Other treatments, including psychotherapeutic interventions, are usually required in addition to antidepressants.

C. Pain Disorders

It has been known for over 40 years that antidepressants possess analgesic properties independent of their mood effects. TCAs have been used in the treatment of neuropathic and other pain conditions since the 1960s. Medications that possess both norepinephrine and 5-HT reuptake blocking properties are often useful in treating pain disorders. Ascending corticospinal monoamine pathways appear to be important in the endogenous analgesic system. In addition, chronic pain conditions are commonly associated with major depression. TCAs continue to be commonly used for some of these conditions, and SNRIs are increasingly used. In 2010, duloxetine was approved for the treatment of chronic joint and muscle pain. As mentioned earlier, milnacipran is approved for the treatment of fibromyalgia in the USA and for MDD in other countries. Other SNRIs, eg, desvenlafaxine, are being investigated for a variety of pain conditions from postherpetic neuralgia to chronic back pain.

D. Premenstrual Dysphoric Disorder

Approximately 5% of women in the child-bearing years will have prominent mood and physical symptoms during the late luteal phase of almost every cycle; these may include anxiety, depressed mood, irritability, insomnia, fatigue, and a variety of other physical symptoms. These symptoms are more severe than those typically seen in premenstrual syndrome (PMS) and can be quite disruptive to vocational and interpersonal activities. The SSRIs are known to be beneficial to many women with PMDD, and fluoxetine and sertraline are approved for this indication. Treating for 2 weeks out of the month in the luteal phase may be as effective as continuous treatment. The rapid effects of SSRIs in PMDD may be associated with rapid increases in pregnenolone levels.

E. Smoking Cessation

Bupropion was approved in 1997 as a treatment for smoking cessation. Approximately twice as many people treated with bupropion as with placebo have a reduced urge to smoke. In addition, patients taking bupropion appear to experience fewer mood symptoms and possibly less weight gain while withdrawing from nicotine dependence. Bupropion appears to be about as effective as nicotine patches in smoking cessation. The mechanism by which bupropion is helpful in this application is unknown, but the drug may mimic nicotine’s effects on dopamine and norepinephrine and may antagonize nicotinic receptors. Nicotine is also known to have antidepressant effects in some people, and bupropion may substitute for this effect.

Other antidepressants may also have a role in the treatment of smoking cessation. Nortriptyline has been shown to be helpful in smoking cessation, but the effects have not been as consistent as those seen with bupropion.

F. Eating Disorders

Bulimia nervosa and anorexia nervosa are potentially devastating disorders. Bulimia is characterized by episodic intake of large amounts of food (binges) followed by ritualistic purging through emesis, the use of laxatives, or other methods. Medical complications of the purging, such as hypokalemia, are common and dangerous. Anorexia is a disorder in which reduced food intake results in a loss of weight of 15% or more of ideal body weight, and the person has a morbid fear of gaining weight and a highly distorted body image. Anorexia is often chronic and may be fatal in 10% or more of cases.

Antidepressants appear to be helpful in the treatment of bulimia but not anorexia. Fluoxetine was approved for the treatment of bulimia in 1996, and other antidepressants have shown benefit in reducing the binge-purge cycle. The primary treatment for anorexia at this time is refeeding, family therapy, and cognitive behavioral therapy.

Bupropion may have some benefits in treating obesity. Nondepressed, obese patients treated with bupropion were able to lose somewhat more weight and maintain the loss relative to a similar population treated with placebo. However, the weight loss was not robust, and there appear to be more effective options for weight loss.

G. Other Uses for Antidepressants

Antidepressants are used for many other on- and off-label applications. Enuresis in children is an older labeled use for some TCAs, but they are less commonly used now because of their side effects. The SNRI duloxetine is approved in Europe for the treatment of urinary stress incontinence. Many of the serotonergic antidepressants appear to be helpful for treating vasomotor symptoms in perimenopause. Desvenlafaxine is under consideration for FDA approval for the treatment of these vasomotor symptoms, and studies have suggested that SSRIs, venlafaxine, and nefazodone may also provide benefit. Although serotonergic antidepressants are commonly associated with inducing sexual adverse effects, some of these effects might prove useful for some sexual disorders. For example, SSRIs are known to delay orgasm in some patients. For this reason, SSRIs are sometimes used to treat premature ejaculation. In addition, bupropion has been used to treat sexual adverse effects associated with SSRI use, although its efficacy for this use has not been consistently demonstrated in controlled trials.

Choosing an Antidepressant

The choice of an antidepressant depends first on the indication. Not all conditions are equally responsive to all antidepressants. However, in the treatment of MDD, it is difficult to demonstrate that one antidepressant is consistently more effective than another. Thus, the choice of an antidepressant for the treatment of depression rests primarily on practical considerations such as cost, availability, adverse effects, potential drug interactions, the patient’s history of response or lack thereof, and patient preference. Other factors such as the patient’s age, gender, and medical status may also guide antidepressant selection. For example, older patients are particularly sensitive to the anticholinergic effects of the TCAs. On the other hand, the CYP3A4-inhibiting effects of the SSRI fluvoxamine may make this a problematic choice in some older patients because fluvoxamine may interact with many other medications that an older patient may require. There is some suggestion that female patients may respond to and tolerate serotonergic better than noradrenergic or TCA antidepressants, but the data supporting this gender difference have not been consistent. Patients with narrow-angle glaucoma may have an exacerbation with noradrenergic antidepressants, whereas bupropion and other antidepressants are known to lower the seizure threshold in epilepsy patients.

At present, SSRIs are the most commonly prescribed first-line agents in the treatment of both MDD and anxiety disorders. Their popularity comes from their ease of use, tolerability, and safety in overdose. The starting dose of the SSRIs is usually the same as the therapeutic dose for most patients, and so titration may not be required. In addition, most SSRIs are now generically available and inexpensive. Other agents, including the SNRIs, bupropion, and mirtazapine, are also reasonable first-line agents for the treatment of MDD. Bupropion, mirtazapine, and nefazodone are the antidepressants with the least association with sexual side effects and are often prescribed for this reason. However, bupropion is not thought to be effective in the treatment of the anxiety disorders and may be poorly tolerated in anxious patients. The primary indication for bupropion is in the treatment of major depression, including seasonal (winter) depression. Off-label uses of bupropion include the treatment of attention deficit hyperkinetic disorder (ADHD), and bupropion is commonly combined with other antidepressants to augment therapeutic response. The primary indication for mirtazapine is in the treatment of major depression. However, its strong antihistamine properties have contributed to its occasional use as a hypnotic and as an adjunctive treatment to more activating antidepressants.

The TCAs and MAOIs are now relegated to second- or third-line treatments for MDD. Both the TCAs and the MAOIs are potentially lethal in overdose, require titration to achieve a therapeutic dose, have serious drug interactions, and have many troublesome adverse effects. As a consequence, their use in the treatment of MDD or anxiety is now reserved for patients who have been unresponsive to other agents. Clearly, there are patients whose depression responds only to MAOIs or TCAs. Thus, TCAs and MAOIs are probably underused in treatment-resistant depressed patients.

The use of antidepressants outside the treatment of MDD tends to require specific agents. For example, the TCAs and SNRIs appear to be useful in the treatment of pain conditions, but other antidepressant classes appear to be far less effective. SSRIs and the highly serotonergic TCA, clomipramine, are effective in the treatment of OCD, but noradrenergic antidepressants have not proved to be as helpful for this condition. Bupropion and nortriptyline have usefulness in the treatment of smoking cessation, but SSRIs have not been proven useful. Thus, outside the treatment of depression, the choice of antidepressant is primarily dependent on the known benefit of a particular antidepressant or class for a particular indication.

Dosing

The optimal dose of an antidepressant depends on the indication and on the patient. For SSRIs, SNRIs, and a number of newer agents, the starting dose for the treatment of depression is usually a therapeutic dose (Table 30–3). Patients who show little or no benefit after at least 4 weeks of treatment may benefit from a higher dose even though it has been difficult to show a clear advantage for higher doses with SSRIs, SNRIs, and other newer antidepressants. The dose is generally titrated to the maximum dosage recommended or to the highest dosage tolerated if the patient is not responsive to lower doses. Some patients may benefit from doses lower than the usual minimum recommended therapeutic dose. TCAs and MAOIs typically require titration to a therapeutic dosage over several weeks. Dosing of the TCAs may be guided by monitoring TCA serum levels.

TABLE 30–3 Antidepressant dose ranges.

image

Some anxiety disorders may require higher doses of antidepressants than are used in the treatment of major depression. For example, patients treated for OCD often require maximum or somewhat higher than maximum recommended MDD doses to achieve optimal benefits. Likewise, the minimum dose of paroxetine for the effective treatment of panic disorder is higher than the minimum dose required for the effective treatment of depression.

In the treatment of pain disorders, modest doses of TCAs are often sufficient. For example, 25–50 mg/d of imipramine might be beneficial in the treatment of pain associated with a neuropathy, but this would be a subtherapeutic dose in the treatment of MDD. In contrast, SNRIs are usually prescribed in pain disorders at the same doses used in the treatment of depression.

Adverse Effects

Although some potential adverse effects are common to all antidepressants, most of their adverse effects are specific to a subclass of agents and to their pharmacodynamic effects. An FDA warning applied to all antidepressants is the risk of increased suicidality in patients younger than 25. The warning suggests that use of antidepressants is associated with suicidal ideation and gestures, but not completed suicides, in up to 4% of patients under 25 who were prescribed antidepressant in clinical trials. This rate is about twice the rate seen with placebo treatment. For those over 25, there is either no increased risk or a reduced risk of suicidal thoughts and gestures on antidepressants, particularly after age 65. Although a small minority of patients may experience a treatment-emergent increase in suicidal ideation with antidepressants, the absence of treatment of a major depressive episode in all age groups is a particularly important risk factor in completed suicides.

A. Selective Serotonin Reuptake Inhibitors

The adverse effects of the most commonly prescribed antidepressants—the SSRIs—can be predicted from their potent inhibition of SERT. SSRIs enhance serotonergic tone, not just in the brain but throughout the body. Increased serotonergic activity in the gut is commonly associated with nausea, gastrointestinal upset, diarrhea, and other gastrointestinal symptoms. Gastrointestinal adverse effects usually emerge early in the course of treatment and tend to improve after the first week. Increasing serotonergic tone at the level of the spinal cord and above is associated with diminished sexual function and interest. As a result, at least 30–40% of patients treated with SSRIs report loss of libido, delayed orgasm, or diminished arousal. The sexual effects often persist as long as the patient remains on the antidepressant but may diminish with time.

Other adverse effects related to the serotonergic effects of SSRIs and vortioxetine include an increase in headaches and insomnia or hypersomnia. Some patients gain weight while taking SSRIs, particularly paroxetine. Sudden discontinuation of short half-life SSRIs such as paroxetine and sertraline is associated with a discontinuation syndrome in some patients characterized by dizziness, paresthesias, and other symptoms beginning 1 or 2 days after stopping the drug and persisting for 1 week or longer.

Most antidepressants are category C agents by the FDA teratogen classification system. There is an association of paroxetine with cardiac septal defects in first trimester exposures. Thus, paroxetine is a category D agent. Other possible associations of SSRIs with post-birth complications, including pulmonary hypertension, have not been clearly established.

B. Serotonin-Norepinephrine Reuptake Inhibitors and Tricyclic Antidepressants

SNRIs have many of the serotonergic adverse effects associated with SSRIs. In addition, SNRIs may also have noradrenergic effects, including increased blood pressure and heart rate, and CNS activation, such as insomnia, anxiety, and agitation. The hemodynamic effects of SNRIs tend not to be problematic in most patients. A dose-related increase in blood pressure has been seen more commonly with the immediate-release form of venlafaxine than with other SNRIs. Likewise, there are more reports of cardiac toxicity with venlafaxine overdose than with either the other SNRIs or SSRIs. Duloxetine is rarely associated with hepatic toxicity in patients with a history of liver damage. All the SNRIs have been associated with a discontinuation syndrome resembling that seen with SSRI discontinuation.

The primary adverse effects of TCAs have been described in the previous text. Anticholinergic effects are perhaps the most common. These effects result in dry mouth, constipation, urinary retention, blurred vision, and confusion. They are more common with tertiary amine TCAs such as amitriptyline and imipramine than with the secondary amine TCAs desipramine and nortriptyline. The potent α-blocking property of TCAs often results in orthostatic hypotension. H1 antagonism by the TCAs is associated with weight gain and sedation. The TCAs are class 1A antiarrhythmic agents (see Chapter 14) and are arrhythmogenic at higher doses. Sexual effects are common, particularly with highly serotonergic TCAs such as clomipramine. The TCAs have a prominent discontinuation syndrome characterized by cholinergic rebound and flulike symptoms.

C. 5-HT Receptor Modulators

The most common adverse effects associated with the 5-HT2 antagonists are sedation and gastrointestinal disturbances. Sedative effects, particularly with trazodone, can be quite pronounced. Thus, it is not surprising that the treatment of insomnia is currently the primary application of trazodone. The gastrointestinal effects appear to be dose-related and are less pronounced than those seen with SNRIs or SSRIs. Sexual effects are uncommon with nefazodone or trazodone treatment as a result of the relatively selective serotonergic effects of these drugs on the 5-HT2 receptor rather than on SERT. However, trazodone has rarely been associated with inducing priapism. Both nefazodone and trazodone are α-blocking agents and may result in a dose-related orthostatic hypotension in some patients. Nefazodone has been associated with hepatotoxicity, including rare fatalities and cases of fulminant hepatic failure requiring transplantation. The rate of serious hepatoxicity with nefazodone has been estimated at 1 in 250,000 to 1 in 300,000 patient-years of nefazodone treatment.

As with the SSRIs, the most common adverse effects of vortioxetine are serotonergic and include dose-dependent gastrointestinal effects, particularly nausea, as well as sexual dysfunction. Higher doses of vortioxetine tend to increase the rate of GI and sexual side effects. The teratogenic risks of vortioxetine are not known but like most other antidepressants, it is considered a category C agent.

D. Tetracyclics and Unicyclics

Amoxapine is sometimes associated with a parkinsonian syndrome due to its D2-blocking action. Mirtazapine has significant sedative effect. Maprotiline has a moderately high affinity for NET and may cause TCA-like adverse effects and, rarely, seizures. Bupropion is occasionally associated with agitation, insomnia, and anorexia. Vilazodone may have somewhat higher rates of gastrointestinal upset, including diarrhea and nausea, than the SSRIs.

E. Monoamine Oxidase Inhibitors

The most common adverse effects of the MAOIs leading to discontinuation of these drugs are orthostatic hypotension and weight gain. In addition, the irreversible nonselective MAOIs are associated with the highest rates of sexual effects of all the antidepressants. Anorgasmia is fairly common with therapeutic doses of some MAOIs. The amphetamine-like properties of some MAOIs contributes to activation, insomnia, and restlessness in some patients. Phenelzine tends to be more sedating than either selegiline or tranylcypromine. Confusion is also sometimes associated with higher doses of MAOIs. Because they block metabolism of tyramine and similar ingested amines, MAOIs may cause dangerous interactions with certain foods and with serotonergic drugs (see Interactions). Finally, MAOIs have been associated with a sudden discontinuation syndrome manifested in a delirium-like presentation with psychosis, excitement, and confusion.

Overdose

Suicide attempts are a common and unfortunate consequence of major depression. The lifetime risk of completing suicide in patients previously hospitalized with MDD may be as high as 15%. Overdose is the most common method used in suicide attempts, and antidepressants, especially the TCAs, are frequently involved. Overdose can induce lethal arrhythmias, including ventricular tachycardia and fibrillation. In addition, blood pressure changes and anticholinergic effects including altered mental status and seizures are sometimes seen in TCA overdoses. A 1500 mg dose of imipramine or amitriptyline (less than 7 days’ supply at antidepressant doses) is enough to be lethal in many patients. Toddlers taking 100 mg will likely show evidence of toxicity. Treatment typically involves cardiac monitoring, airway support, and gastric lavage. Sodium bicarbonate is often administered to displace the TCA from cardiac sodium channels.

An overdose with an MAOI can produce a variety of effects including autonomic instability, hyperadrenergic symptoms, psychotic symptoms, confusion, delirium, fever, and seizures. Management of MAOI overdoses usually involves cardiac monitoring, vital signs support, and lavage.

Compared with TCAs and MAOIs, the other antidepressants are generally much safer in overdose. Fatalities with SSRI overdose alone are extremely uncommon. Similarly, SNRIs tend to be much safer in overdose than the TCAs. However, venlafaxine has been associated with some cardiac toxicity in overdose and appears to be less safe than SSRIs. Bupropion is associated with seizures in overdose, and mirtazapine may be associated with sedation, disorientation, and tachycardia. With the newer agents, fatal overdoses often involve the combination of the antidepressant with other drugs, including alcohol. Management of overdose with the newer antidepressants usually involves emptying of gastric contents and vital sign support as the initial intervention.

Drug Interactions

Antidepressants are commonly prescribed with other psychotropic and nonpsychotropic agents. There is potential for drug interactions with all antidepressants, but the most serious of these involve the MAOIs and to a lesser extent the TCAs.

A. Selective Serotonin Reuptake Inhibitors

The most common interactions with SSRIs are pharmacokinetic interactions. For example, paroxetine and fluoxetine are potent CYP2D6 inhibitors (Table 30–4). Thus, administration with 2D6 substrates such as TCAs can lead to dramatic and sometimes unpredictable elevations in the tricyclic drug concentration. The result may be toxicity from the TCA. Similarly, fluvoxamine, a CYP3A4 inhibitor, may elevate the levels of concurrently administered substrates for this enzyme such as diltiazem and induce bradycardia or hypotension. Other SSRIs, such as citalopram and escitalopram, are relatively free of pharmacokinetic interactions. The most serious interaction with the SSRIs are pharmacodynamic interactions with MAOIs that produce a serotonin syndrome (see below).

TABLE 30–4 Some antidepressant–CYP450 drug interactions.

image

B. Selective Serotonin-Norepinephrine Reuptake Inhibitors and Tricyclic Antidepressants

The SNRIs have relatively fewer CYP450 interactions than the SSRIs. Venlafaxine is a substrate but not an inhibitor of CYP2D6 or other isoenzymes, whereas desvenlafaxine is a minor substrate for CYP3A4. Duloxetine is a moderate inhibitor of CYP2D6 and so may elevate TCA and levels of other CYP2D6 substrates. Since milnacipran is neither a substrate nor potent inducer of CYP450 isoenzymes, is not tightly protein bound, and is largely excreted unchanged in the urine, it is unlikely to have clinically significant pharmacokinetic drug interactions. On the other hand, levomilnacipran is reported to be a substrate of CYP3A4 and the dosage of the drug should be lowered when combined with potent inhibitors of CYP3A4 such as ketoconazole. Like all serotonergic antidepressants, SNRIs are contraindicated in combination with MAOIs.

Elevated TCA levels may occur when these drugs are combined with CYP2D6 inhibitors or from constitutional factors. About 7% of the Caucasian population in the USA has a CYP2D6 polymorphism that is associated with slow metabolism of TCAs and other 2D6 substrates. Combination of a known CYP2D6 inhibitor and a TCA in a patient who is a slow metabolizer may result in markedly increased effects. Such an interaction has been implicated, though rarely, in cases of TCA toxicity. There may also be additive anticholinergic or antihistamine effects when TCAs are combined with other agents that share these properties such as benztropine or diphenhydramine. Similarly, antihypertensive drugs may exacerbate the orthostatic hypotension induced by TCAs.

C. 5-HT Receptor Modulators

Nefazodone is an inhibitor of the CYP3A4 isoenzyme, so it can raise the level and thus exacerbate adverse effects of many 3A4-dependent drugs. For example, triazolam levels are increased by concurrent administration of nefazodone such that a reduction in triazolam dosage by 75% is recommended. Likewise, administration of nefazodone with simvastatin has been associated with 20-fold increase in plasma levels of simvastatin.

Trazodone is a substrate but not a potent inhibitor of CYP3A4. As a result, combining trazodone with potent inhibitors of CYP3A4, such as ritonavir or ketoconazole, may lead to substantial increases in trazodone levels.

Vortioxetine is a substrate of CYP2D6 and 2B6 and it is recommended that the dose be cut in half when it is co-administered with fluoxetine or bupropion. Inducers of CYP isoenzymes such as rifampin, carbamazepine, and phenytoin will lower serum levels of vortioxetine and may require increasing the dose of vortioxetine.

D. Tetracyclic and Unicyclic Antidepressants

Bupropion is metabolized primarily by CYP2B6, and its metabolism may be altered by drugs such as cyclophosphamide, which is a substrate of 2B6. The major metabolite of bupropion, hydroxybupropion, is a moderate inhibitor of CYP2D6 and so can raise desi-pramine levels. Bupropion should be avoided in patients taking MAOIs.

Mirtazapine is a substrate for several CYP450 enzymes including 2D6, 3A4, and 1A2. Consequently, drugs that inhibit these isozymes may raise mirtazapine levels. However, mirtazapine is not an inhibitor of these enzymes. The sedating effects of mirtazapine may be additive with those of CNS depressants such as alcohol and benzodiazepines.

Amoxapine and maprotiline share most drug interactions common to the TCA group. Both are CYP2D6 substrates and should be used with caution in combination with inhibitors such as fluoxetine. Amoxapine and maprotiline also both have anticholinergic and antihistaminic properties that may be additive with drugs that share a similar profile.

Since vilazodone is primarily a substrate of CYP3A4, strong CYP3A4 inhibitors such as ketoconazole can increase the serum concentration of vilazodone by 50% or more. On the other hand, vilazodone is neither a potent inhibitor nor a strong inducer of any CYP isoenzymes. It may be a mild inducer of CYP2C19.

E. Monoamine Oxidase Inhibitors

MAOIs are associated with two classes of serious drug interactions. The first of these is the pharmacodynamic interaction of MAOIs with serotonergic agents including SSRIs, SNRIs, and most TCAs along with some analgesic agents such as meperidine. These combinations of an MAOI with a serotonergic agent may result in a life-threatening serotonin syndrome (see Chapter 16). The serotonin syndrome is thought to be caused by overstimulation of 5-HT receptors in the central gray nuclei and the medulla. Symptoms range from mild to lethal and include a triad of cognitive (delirium, coma), autonomic (hypertension, tachycardia, diaphoreses), and somatic (myoclonus, hyperreflexia, tremor) effects. Most serotonergic antidepressants should be discontinued at least 2 weeks before starting an MAOI. Fluoxetine, because of its long half-life, should be discontinued for 4–5 weeks before an MAOI is initiated. Conversely, an MAOI must be discontinued for at least 2 weeks before starting a serotonergic agent.

The second serious interaction with MAOIs occurs when an MAOI is combined with tyramine in the diet or with sympathomimetic substrates of MAO. An MAOI prevents the breakdown of tyramine in the gut, and this results in high serum levels that enhance peripheral noradrenergic effects, including raising blood pressure dramatically. Patients on an MAOI who ingest large amounts of dietary tyramine may experience malignant hypertension and subsequently a stroke or myocardial infarction. Thus, patients taking MAOIs require a low-tyramine diet and should avoid foods such as aged cheeses, tap beer, soy products, and dried sausages, which contain high amounts of tyramine (see Chapter 9). Similar sympathomimetics also may cause significant hypertension when combined with MAOIs. Thus, over-the-counter cold preparations that contain pseudoephedrine and phenylpropanolamine are contraindicated in patients taking MAOIs.

SUMMARY Antidepressants

image

PREPARATIONS AVAILABLE

image

image

REFERENCES

Aan Het Rot M et al: Ketamine for depression: Where do we go from here? Biol Psychiatry 2012;72:537.

Alam MY et al: Safety, tolerability, and efficacy of vortioxetine (Lu AA21004) in major depressive disorder: Results of an open-label, flexible-dose, 52-week extension study. Int Clin Psychopharmacol 2014;29:39.

Alessandro S, Kato M: The serotonin transporter gene and effectiveness of SSRIs. Expert Rev Neurother 2008;8(1):111.

Bab I, Yirmiya R: Depression, selective serotonin reuptake inhibitors, and osteoporosis. Curr Osteoporos Rep 2010;8:185.

Barrera AZ, Torres LD, Munoz RF: Prevention of depression: The state of the science at the beginning of the 21st century. Int Rev Psychiatry 2007;19:655.

Bellingham GA, Peng PW: Duloxetine: A review of its pharmacology and use in chronic pain management. Reg Anesth Pain Med 2010;35:294.

Belmaker R, Agam G: Major depressive disorder. N Engl J Med 2008;358:55.

Bockting CL et al: Continuation and maintenance use of antidepressants in recurrent depression. Psychother Psychosom 2008;77:17.

Bonisch H, Bruss M: The norepinephrine transporter in physiology and disease. Handb Exp Pharmacol 2006;175:485.

Castren E, Voikar V, Rantamaki T: Role of neurotrophic factors in depression. Curr Opin Pharmacol 2007;7:18.

Chaki S et al: mGlu2/3 and mGlu5 receptors: Potential targets for novel antidepressants. Neuropharmacology 2013;66:40.

Chappell AS et al: A double-blind, randomized, placebo-controlled study of the efficacy and safety of duloxetine for the treatment of chronic pain due to osteoarthritis of the knee. Pain Pract 2011;11:33.

Chen G et al: Pharmacokinetic drug interactions involving vortioxetine (Lu AA21004), a multimodal antidepressant. Clin Drug Investig 2013;33:727.

Cipriani A et al: Fluoxetine versus other types of pharmacotherapy for depression. Cochrane Database Syst Rev 2005;(4):CD004185.

Cipriani A et al: Metareview on short-term effectiveness and safety of antidepressants for depression: An evidence-based approach to inform clinical practice. Can J Psychiatry 2007;52:553.

Citrome L: Vortioxetine for major depressive disorder: A systematic review of the efficacy and safety profile for this newly approved antidepressant - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract 2014;68:60.

de Beaurepaire R: Questions raised by the cytokine hypothesis of depression. Brain Behav Immun 2002;16:610.

Dhillon S, Scott LJ, Plosker GL: Escitalopram: A review of its use in the management of anxiety disorders. CNS Drugs 2006;20:763.

Dhillon S, Yang LP, Curran MP: Bupropion: A review of its use in the management of major depressive disorder. Drugs 2008;68:653.

Duman RS, Monteggia LM: A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006;59:1116.

Dvir Y, Smallwood P: Serotonin syndrome: A complex but easily avoidable condition. Gen Hosp Psychiatry 2008;30:284.

Fontenelle LF et al: An update on the pharmacological treatment of obsessive-compulsive disorder. Expert Opin Pharmacother 2007;8:563.

Geisser ME et al: A pooled analysis of two randomized, double-blind, placebo-controlled trials of milnacipran monotherapy in the treatment of fibromyalgia. Pain Pract 2011;11:120.

Gether U et al: Neurotransmitter transporters: Molecular function of important drug targets. Trends Pharmacol Sci 2006;27:375.

Gillespie CF, Nemeroff CB: Hypercortisolemia and depression. Psychosom Med 2005;67(Suppl 1):S26.

Gillman PK: A review of serotonin toxicity data: Implications for the mechanisms of antidepressant drug action. Biol Psychiatry 2006;59:1046.

Gillman PK: Tricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br J Pharmacol 2007;151:737.

Giner L et al: Selective serotonin reuptake inhibitors and the risk for suicidality in adolescents: An update. Int J Adolesc Med Health 2005;17:211.

Guay DR: Vilazodone hydrochloride, a combined SSRI and 5-HT1A receptor agonist for major depressive disorder. Consult Pharm 2012;27:857.

Gutman DA, Owens MJ: Serotonin and norepinephrine transporter binding profile of SSRIs. Essent Psychopharmacol 2006;7:35.

Hirschfeld RM: Antidepressants in long-term therapy: A review of tricyclic antidepressants and selective serotonin reuptake inhibitors. Acta Psychiatr Scand Suppl 2000;403:35.

Hirschfeld RM: History and evolution of the monoamine hypothesis of depression. J Clin Psychiatry 2000;61(Suppl 6):4.

Holma KM et al: Long-term outcome of major depressive disorder in psychiatric patients is variable. J Clin Psychiatry 2008;69:196.

Jann MW, Slade JH: Antidepressant agents for the treatment of chronic pain and depression. Pharmacotherapy 2007;27:1571.

Kalia M: Neurobiological basis of depression: An update. Metabolism 2005;54(5 Suppl 1):24.

Kozisek ME, Middlemas D, Bylund DB: Brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B in the mechanism of action of antidepressant therapies. Pharmacol Ther 2008;117:30.

Krystal JH, Sanacora G, Duman RS. Rapid-acting glutamatergic antidepressants: The path to ketamine and beyond. Biol Psychiatry 2013;73:1133.

Laughren TP et al: Vilazodone: Clinical basis for the US Food and Drug Administration’s approval of a new antidepressant. J Clin Psychiatry 2011;72:1166.

Lesch KP, Gutknecht L: Pharmacogenetics of the serotonin transporter. Prog Neuropsychopharmacol Biol Psychiatry 2005;29:1062.

Mago R et al: Safety and tolerability of levomilnacipran ER in major depressive disorder: Results from an open-label, 48-week extension study. Clin Drug Investig 2013;33:761.

Maletic V et al: Neurobiology of depression: An integrated view of key findings. Int J Clin Pract 2007;61:2030.

Manji HK, Drevets WC, Charney DS: The cellular neurobiology of depression. Nat Med 2001;7:541.

Mathews DC, Zarate CA Jr. Current status of ketamine and related compounds for depression. J Clin Psychiatry 2013;74:516.

McCleane G: Antidepressants as analgesics. CNS Drugs 2008;22:139.

McEwen BS: Glucocorticoids, depression, and mood disorders: Structural remodeling in the brain. Metabolism 2005;54(5 Suppl 1):20.

Moller HJ: Evidence for beneficial effects of antidepressants on suicidality in depressive patients: A systematic review. Eur Arch Psychiatry Clin Neurosci 2006;256:329.

Montgomery SA, et al: Efficacy and safety of levomilnacipran sustained release in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2013;74:363.

Nestler EJ et al: Neurobiology of depression. Neuron 2002;34:13.

Pace TW, Hu F, Miller AH: Cytokine-effects on glucocorticoid receptor function: Relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun 2007;21:9.

Pilc A, Wieronska JM, Skolnick P: Glutamate-based antidepressants: Preclinical psychopharmacology. Biol Psychiatry 2013;73:1125.

Sakinofsky I: Treating suicidality in depressive illness. Part 2: Does treatment cure or cause suicidality? Can J Psychiatry 2007;52(6 Suppl 1):85S.

Schatzberg AF, Cole JO, DeBattista C: Manual of Clinical Psychopharmacology, 6th ed. American Psychiatric Publishing, 2007.

Shapiro JR et al: Bulimia nervosa treatment: A systematic review of randomized controlled trials. Int J Eat Disord 2007;40:321.

Soomro GM et al: Selective serotonin reuptake inhibitors (SSRIs) versus placebo for obsessive compulsive disorder (OCD). Cochrane Database Syst Rev 2008;(1):CD001765.

Stein MB, Stein DJ: Social anxiety disorder. Lancet 2008;371(9618):1115.

Stone EA, Lin Y, Quartermain D: A final common pathway for depression? Progress toward a general conceptual framework. Neurosci Biobehav Rev 2008;32:508.

Tuccori M et al: Use of selective serotonin reuptake inhibitors during pregnancy and risk of major and cardiovascular malformations: An update. Postgrad Med 2010;122:49.

Warden D et al: The STAR*D Project results: A comprehensive review of findings. Curr Psychiatry Rep 2007;9:449.

Wheeler BW et al: The population impact on incidence of suicide and non-fatal self harm of regulatory action against the use of selective serotonin reuptake inhibitors in under 18s in the United Kingdom: Ecological study. Br Med J 2008;336(7643):542.

Wilson KL et al: Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol 2011;28:19.

Yu S, Holsboer F, Almeida OF: Neuronal actions of glucocorticoids: Focus on depression. J Steroid Biochem Mol Biol 2008;108:300.

CASE STUDY ANSWER

Fluoxetine, the prototype SSRI, has a number of pharmacokinetic and pharmacodynamic interactions. Fluoxetine is a CYP450 2D6 inhibitor and thus can inhibit the metabolism of 2D6 substrates such as propranolol and other β blockers; tricyclic antidepressants; tramadol; opioids such as methadone, codeine, and oxycodone; antipsychotics such as haloperidol and thioridazine; and many other drugs. This inhibition of metabolism can result in significantly higher plasma levels of the concurrent drug, and this may lead to an increase in adverse reactions associated with that drug. As a potent inhibitor of the serotonin transporter, fluoxetine is associated with a number of pharmacodynamic interactions involving serotonergic neurotransmission. The combination of tramadol with fluoxetine has occasionally been associated with a serotonin syndrome, characterized by diaphoreses, autonomic instability, myoclonus, seizures, and coma. The combination of fluoxetine with an MAOI is contraindicated because of the risk of a fatal serotonin syndrome. In addition, meperidine is specifically contraindicated in combination with an MAOI.



If you find an error or have any questions, please email us at admin@doctorlib.org. Thank you!