National Cancer Institute
U.S. National Institutes of Health | www.cancer.gov

NCI Home
Cancer Topics
Clinical Trials
Cancer Statistics
Research & Funding
News
About NCI
Depression (PDQ®)
Patient Version   Health Professional Version   En español   Last Modified: 12/19/2008



Purpose of This PDQ Summary






Overview






Assessment and Diagnosis






Intervention






Suicide Risk in Cancer Patients






Assessment, Evaluation, and Management of Suicidal Patients






Pediatric Considerations for Depression






Pediatric Considerations for Suicidality






Get More Information From NCI






Changes to This Summary (12/19/2008)






Questions or Comments About This Summary






More Information



Page Options
Print This Page
Print Entire Document
View Entire Document
E-Mail This Document
Quick Links
Director's Corner

Dictionary of Cancer Terms

NCI Drug Dictionary

Funding Opportunities

NCI Publications

Advisory Boards and Groups

Science Serving People

Español
Quit Smoking Today
NCI Highlights
Report to Nation Finds Declines in Cancer Incidence, Death Rates

High Dose Chemotherapy Prolongs Survival for Leukemia

Prostate Cancer Study Shows No Benefit for Selenium, Vitamin E

The Nation's Investment in Cancer Research FY 2009

Past Highlights
Intervention

Pharmacologic Intervention
        Overview
        Suicide risk of antidepressant medication
        Interferon-related depression
        Antidepressant medication selection
        Selective serotonin reuptake inhibitors
        Discontinuation of antidepressants
        Side effects
        Drug-drug interactions
        Atypical antidepressants
        Benzodiazepines
        Psychostimulants
        Monoamine oxidase inhibitors
        St. John's wort
        Antidepressant effects
Psychotherapy
        Overview
        Empirical studies of the efficacy of psychotherapy

Whether to initiate therapy for depression depends on the probability that the patient will recover spontaneously in the next 2 to 4 weeks, the degree of functional impairment, and the severity and duration of the depressive symptoms.[1] Studies have shown that treatment of major depression is optimized by a combination of pharmacotherapy and psychotherapy. Thus, even if a primary care physician or oncologist undertakes the treatment of depressive symptoms pharmacologically, a referral for psychotherapy or supportive counseling should be considered.

Individuals should be referred for a psychiatric consultation for the following reasons:

  • A primary care physician or oncologist does not feel competent treating the patient for depression because of specific clinical features in the presentation (i.e., if prominent suicidal tendencies are present).


  • The depressive symptoms treated by the primary physician are resistant to pharmacologic interventions after 2 to 4 weeks of intervention.


  • The depressive symptoms are worsening rather than improving.


  • Initiating treatment with antidepressant drugs, titrating drug doses, or continuing treatment is interrupted or made problematic by adverse effects attributable to the medication.


  • The depressive symptoms are interfering with the patient's ability to be cooperative with medical treatment.[2-4]


Pharmacologic Intervention

Overview

There is a paucity of randomized, placebo-controlled trials assessing the risks and benefits of antidepressants in patients with cancer and depression or depressive symptoms. Furthermore, these studies are limited by methodological challenges and a lack of broad representation of children, adolescents, older adults, and minority groups.[5] In certain cases of depression in patients with cancer, antidepressant therapy may be indicated. A survey of prescribing patterns in outpatient oncology settings over a 2-year period found that antidepressants were prescribed for about 14% of patients.[6] In a systematic review of newer pharmacotherapies for depression in adults, the response rate for treatment of depression with antidepressants was found to be approximately 54%.[7] The efficacy of the newer pharmacotherapies is similar to that of older antidepressants for general medical patients, including older adults and those with coexisting medical or psychiatric illness.[7] The dropout rates due to adverse effects are approximately 11% for newer antidepressants and 16% for older antidepressants.[7] Because of the relative paucity of data regarding antidepressant use in oncology settings, there is considerable variability in practice patterns related to prescribing antidepressants in cancer patients. Although studies generally indicate that about 25% of all cancer patients are depressed, one study found that only 16% of cancer patients were receiving antidepressant medication.[8]

Antidepressant Studies

  • In adults, a double-blind placebo-controlled trial comparing fluoxetine with desipramine in treating depressive symptoms in 40 women with cancer found both medications to be effective and well tolerated. There were greater improvements on several quality-of-life measures in patients who received fluoxetine.[9]


  • One study compared paroxetine with amitriptyline in the management of depression in women with breast cancer. Both treatments were equally effective. Paroxetine was associated with significantly fewer anticholinergic adverse effects than amitriptyline.[10]


  • In a randomized controlled trial comparing fluoxetine with a placebo, patients receiving fluoxetine were found to have improved quality of life and decreased depressive symptoms.[11] Using a symptom-based approach (similar to the management of other cancer-related symptoms such as pain or nausea), this study assessed for depression by use of a 2-item screening procedure focused on presence of anhedonia (little interest or pleasure in doing things) and depressed or hopeless mood. Most of the sample consisted of patients with mild-to-moderate levels of depressive symptoms regardless of whether they met the diagnostic criteria for depression. Generalization was enhanced by inclusion of a sample of mixed cancer types (e.g., breast, thoracic, genitourinary, gastrointestinal) from a predominantly community cancer care setting, an equal male/female ratio, and a relatively large sample size (n = 163). A subgroup of patients identified as having higher levels of depressive symptoms was most likely to benefit from the treatment.


Suicide risk of antidepressant medication

Over the past few years, significant concerns have been raised about the risk of suicidal thinking and behavior with the use of antidepressants in children, adolescents, and young adults. Since 2003, U.S. and European regulators have issued several public health warnings on this topic. The first such advisory issued by the U.S. Food and Drug Administration (FDA) warned about a possible association between antidepressants and suicidal thinking and behavior in children and adolescents. In December 2003, the Medicines and Healthcare Products Regulatory Agency of the United Kingdom issued a letter to doctors advising against the use of antidepressants in anyone younger than 18 years.[12] In October 2004, the FDA mandated pharmaceutical companies to add a “black box” warning to the labeling of all antidepressants suggesting increased risk of suicidality in pediatric patients who were taking antidepressants. The FDA revised this black box warning in May 2007 to include young adults younger than 25 years.[13] The new, carefully worded warning emphasizes that the risk of suicidality is associated with both antidepressants and depression. In addition to raising concerns about increased sucidality in children, adolescents, and young adults, the warning acknowledges a significant protective effect of antidepressants in adults aged 65 and older. The meta-analysis that led to the initial black box warning in pediatric patients concluded that the antidepressants are associated with a twofold increase in suicidal ideation and behavior compared to the placebo in children and adolescents. A major meta-analysis published in the Journal of the American Medical Association reanalyzed the data from the child and adolescent studies (including seven studies not included in the initial meta-analysis), using a random-effects model. While this reanalysis found an overall increased risk of suicidal ideation/suicidal behavior consistent with the initial meta-analysis, the pooled risk differences were found to be smaller and statistically insignificant. Concerns have been raised that the unintended consequence of the warnings will be overly restricted use of antidepressants among those who benefit the most and, hence, an increase in suicidality that the warning seeks to prevent. A study examining U.S. and Dutch data suggests a drop in selective serotonin reuptake inhibitor (SSRI) prescriptions for children and adolescents since the black box warning was issued and a simultaneous increase in suicide rates in this patient population. In summary, the risk-benefit equation favors appropriate use of antidepressants with careful monitoring for suicidality. It is important to note that none of the studies that led to the black box warning included or focused on patients being treated for cancer. Clinical experience and results of small clinical trials suggest that antidepressants can be safely administered to adult cancer patients, although there are no large controlled clinical trials to support this position. When antidepressants are prescribed for patients with cancer, a careful monitoring plan should be implemented by individuals with expertise, and consultation referral should be made for patients who do not respond as anticipated or who present other concerns.

Interferon-related depression

Most antidepressant prescribing is directed at the treatment of an existing depressive disorder or significant depressive symptoms. One study, however, supports the use of antidepressants to prevent depression in patients receiving high-dose interferon for adjuvant therapy of malignant melanoma.[14] The rationale for this approach is that treatment with high-dose interferon is associated with a particularly high rate of depression in this patient population, and proinflammatory cytokines implicated in the biological changes that result in depression may be directly reduced by antidepressants. In this double-blind study of patients receiving high-dose interferon, 2 of 18 patients in the paroxetine group developed depression during the first 12 weeks of therapy, compared with 9 of 20 patients in the placebo group (relative risk [RR] = 0.24; 95% confidence interval [CI], 0.08–0.93). Moreover, there were significantly fewer treatment discontinuations in the paroxetine group (5% vs. 35%, RR = 0.14; 95% CI, 0.05–0.85). Further study is required to confirm these findings and to determine whether prophylactic use of antidepressants has benefit in other treatment settings.

Antidepressant medication selection

The choice of antidepressant depends on a patient's medical history and concomitant medical problems, the symptoms referable to depression, previous responses to antidepressant medications, and the side effects associated with the agents available.

The types of medications used to treat depression in patients with cancer include the SSRIs, tricyclic antidepressants (TCAs), and analeptic or CNS stimulant agents (i.e., amphetamines). The following table outlines the commonly used antidepressants and highlights starting dosages used in cancer patients. The Side Effects/Comments column identifies drug-specific side effects that may be clinically advantageous or problematic depending on the clinical situation when selecting antidepressant medications and monitoring patients receiving these drugs. Generally, there is a long latency period (3–6 weeks) from initiation of antidepressant medications until the onset of a therapeutic response. In many cases, antidepressant treatment begins at low doses followed by a period of gradual dose titration to achieve an optimum individualized response. Initial low doses may help to avoid initial side effects, but dose escalation may be required in order to see therapeutic effects. For some agents, there is a therapeutic window during which plasma concentrations correlate with a patient's antidepressant response (e.g., nortriptyline). For patients receiving these agents, serial drug concentration monitoring guides therapy and facilitates providing an adequate therapeutic trial, because plasma concentrations less than and greater than the defined therapeutic ranges are associated with treatment failure, suboptimal responses, and in the case of high drug concentrations, unnecessary toxicity.

Antidepressant Medications for Ambulatory Adult Patients
*Drug Class/Generic Name (Proprietary Name)/Dosages  Side Effects/Comments 
TRICYCLIC ANTIDEPRESSANTS (TCAs) All TCAs can cause cardiac arrhythmias.
EKG at baseline to evaluate for preexisting cardiac conduction abnormalities. Therapeutic drug concentration ranges in plasma have been identified for all agents, but dosage adjustments should be based on a patient's clinical response and not solely on plasma concentrations.a
In responding patients, decrease daily dosages to the lowest effective amount needed to sustain a response.b TCAs can cause sexual dysfunction.
Treatment may be associated with weight gain.c
amitriptyline (Elavil) Marked sedation; dizziness; headache; weight gain; anticholinergic effects;d orthostatic blood pressure (BP) changes (postural hypotension); may produce sexual dysfunction. Therapeutic plasma concentrations (parent drug + active metabolite) = 110–250 ng/mL.
initial: 10–25 mg as a single daily dose, preferably at bedtime
maintenance: 150–300 mg/day
clomipramine (Anafranil) Anticholinergic effects; dizziness; drowsiness; headache; weight gain; orthostatic hypotension.
initial: 25 mg/day and gradually increase to 100 mg/day the first 2 weeks; may be given at bedtime
maintenance: 100–250 mg/day maximum
desipramine (Norpramin) Mild sedation; increased appetite; nausea; minimal anticholinergic effects;d orthostatic BP changes. Therapeutic plasma concentrations = 125–300 ng/mL.
initial: 25–50 mg/day as a single daily dose, preferably at bedtime
maintenance: 100–300 mg/day as a single daily dose; In elderly patients, daily doses >150 mg are not recommended
doxepin (Sinequan) Moderately to very sedating; dizziness; headache; weight gain; moderate anticholinergic effects;d postural hypotension. Optimal antidepressant effect is characteristically delayed by 2–3 weeks; however, onset of antianxiety effect is comparatively rapid. Therapeutic plasma concentrations (parent drug + active metabolite) = 100–200 ng/mL.
initial: 10–25 mg/day as a single daily dose, preferably at bedtime
maintenance: 75–300 mg/day as a single daily dose, preferably at bedtime
imipramine (Tofranil) Moderately to very sedating; dizziness; headache; weight gain; moderate anticholinergic effects;d moderate-marked orthostatic BP changes; may produce sexual dysfunction (both genders). Therapeutic plasma concentrations (parent drug + active metabolite) = 200–350 ng/mL.
initial: 25–50 mg/day as a single daily dose, preferably at bedtime
maintenance: 75–200 mg/day as a single daily dose, preferably at bedtime
nortriptyline (Pamelor, Aventyl) Mild-moderate sedation; constipation; nausea; increased appetite; mild-moderate anticholinergic effects;d the TCA least likely to produce postural hypotension. Therapeutic plasma concentrations = 50–150 ng/mL.
initial: 10–25 mg, 3–4 times daily
maintenance: 30–50 mg, 3 times daily, daily doses >150 mg are not recommended
SELECTIVE SEROTONIN REUPTAKE INHIBITORS (SSRIs) SSRIs have few anticholinergic and cardiovascular adverse effects. Life-threatening and fatal reactions have occurred in patients who receive SSRIs within 2 weeks of using monoamine oxidase inhibitor antidepressants. Sexual dysfunction has been reported to be associated with SSRI use. There is limited experience with long-term use.
citalopram (Celexa) Ejaculation disorder; other sexual dysfunctions; insomnia; dry mouth; nausea; somnolence. In vitro studies indicated that CYP3A4 and CYP2C19 are the primary enzymes involved in the metabolism of citalopram. Citalopram is a relatively weak inhibitor of CYP2D6.
initial: 10 mg/day
maintenance: 10–40 mg/day
fluoxetine (Prozac) Anxiety; nervousness; insomnia; anorexia; mild bradycardia; sinoatrial node slowing; weight loss; solar photosensitivity; hyponatremia; sexual dysfunction; may alter glycemic control in diabetic patients. Fluoxetine substantially inhibits CYP2D6 and may inhibit the clearance of other drugs metabolized by cytochrome P450 CYP2D6 isozymes.[15] Fluoxetine probably inhibits CYP2C9/10, moderately inhibits CYP2C19, and mildly inhibits CYP3A4;[15] fluoxetine metabolism is impaired in elderly patients.
initial: 10–20 mg/day
maintenance: 20–80 mg/day
escitalopram (Lexapro) Nausea, vomiting, diarrhea, constipation, upset stomach, loss of appetite, dizziness, drowsiness, trouble sleeping, back pain, or dry mouth.
initial: 10 mg/day
maintenance: 10–20 mg/day
fluvoxamine (Luvox) Nausea; sexual dysfunction; headache; nervousness; insomnia; drowsiness.
initial: 50 mg at bedtime, adjust in 50 mg increments at 4- to 7-day intervals
maintenance: 100–300 mg/day
paroxetine (Paxil) Anxiety; nervousness; insomnia; mild weight loss; headache; solar photosensitivity; hyponatremia; sexual dysfunction. Paroxetine substantially inhibits and may interact with other drugs metabolized by cytochrome P450 CYP2D6 isozyme.[15] Paroxetine metabolism is impaired in elderly patients.
initial: 10–20 mg/day
maintenance: 20–50 mg/day
sertraline (Zoloft) Anxiety; nervousness; insomnia; mild weight loss; headache; solar photosensitivity; hyponatremia; sexual dysfunction. Produces mild inhibition of and may interact with drugs metabolized by cytochrome P450 CYP2D6 isozymes with little, if any, effect on CYP1A2, CYP2C9/10, CYP2C19, or CYP3A3/4.[15]
initial: 25–50 mg/day
maintenance: 50–200 mg/day
MONOAMINE OXIDASE INHIBITORS (MAOIs)
tranylcypromine (Parnate) Orthostatic hypotension; drowsiness; hyperexcitability; headache. Low tyramine diet required.
initial: 10 mg twice daily, increase by 10 mg increments at 1- to 3-week intervals
maintenance: 10–40 mg/day
phenelzine (Nardil) Orthostatic hypotension; drowsiness; hyperexcitability; headache. Low tyramine diet required.
initial: 15 mg 3 times a day
maintenance: 15–90 mg/day
selegiline (EMSAM) Application site reaction; orthostatic hypotension; diarrhea; headache; insomnia; dry mouth. Any dosages higher than 6 mg/24 h require low-tyramine diet.
initial: 6-mg patch/24 h (20-mg patch topically every 24 h)
maintenance: 6-mg patch/24 h (20-mg patch topically every 24 h). May increase at increments of 3 mg/24 h at 2-week intervals up to 12 mg/24 h.
ATYPICAL ANTIDEPRESSANTS In general, serum drug concentrations do not correlate with antidepressant response.
bupropion (Wellbutrin, also approved for the treatment of smoking cessation as Zyban) Initially activating dose-related seizure-inducing potential; contraindicated in patients with CNS involvement, with a history of seizure, in those with concomitant conditions predisposing to seizure, and in patients taking other drugs that lower seizure threshold. Mild-moderate sedation; mild-moderate anticholinergic effects;d mild orthostatic BP changes; agitation; insomnia; headache; confusion; dizziness; seizures; weight loss.
initial: 75 mg/day
maintenance: 200–450 mg/day not to exceed 150 mg/dose
trazodone (Desyrel) Mild-moderate sedation; negligible anticholinergic effects; mild-moderate orthostatic BP changes, particularly in elderly patients; dizziness; headache; confusion; muscle tremors; may produce priapism; taking trazodone with food can decrease gastrointestinal upset. Therapeutic plasma concentrations = 800–1,600 ng/mL.
initial: 50 mg/day
maintenance: 150–600 mg/day
nefazodone (Serzone) Postural hypotension (although <TCAs); less sexual dysfunction than is reported with SSRIs. Headache; drowsiness; insomnia; agitation; confusion; nausea; tremor. Potential interaction with drugs metabolized by cytochrome P450 isozymes, CYP2D6 and CYP3A4. Check liver function tests at baseline and periodically during therapy. May cause fatal hepatotoxicity.
initial: 100 mg twice daily
maintenance: 300–600 mg/day
mirtazapine (Remeron) A tetracyclic antidepressant. Mirtazapine elimination is decreased in elderly persons. Somnolence; dizziness; increased appetite and weight gain; constipation; hypertension; edema; confusion; increased nonfasting triglycerides and cholesterol; significantly increased hepatic ALT; orthostatic hypotension. When used concomitantly with drugs that reduce the seizure threshold (e.g., phenothiazines), mirtazapine may increase the risk of seizure.
initial: 7.5–15 mg/day
maintenance: 15–45 mg/day
venlafaxine (Effexor) Dose-related sustained hypertension. Headache; dizziness; insomnia; nausea; constipation; abnormal ejaculation. Life-threatening and fatal reactions have occurred in patients who received venlafaxine within 2 weeks of using monoamine oxidase antidepressants.
initial: 75 mg/day
maintenance: 150–375 mg/day
duloxetine (Cymbalta) Nausea, dry mouth, constipation, decreased appetite, fatigue, sleepiness, and increased sweating; decreased sexual drive or ability; urinary hesitation.
initial: 30 mg/day
maintenance: 30–60 mg/day
Psychostimulants Psychostimulants may cause restlessness, agitation, insomnia, nightmares, psychosis, anorexia; and may exacerbate preexisting cardiac disease. Psychostimulants should be administered early in a patient's daily waking cycle. Psychostimulants are sometimes used adjuvantly to antagonize opioid analgesics' sedative effects.
dextroamphetamine (Dexedrine) Drug tolerance, abuse, and dependence liability. Arrhythmia; nervousness; restlessness; insomnia. Contraindicated in patients with advanced arteriosclerosis, symptomatic cardiovascular disease, moderate-severe hypertension, and glaucoma.
initial: 2.5–5 mg/day
maintenance: 10–30 mg/day
methylphenidate (Ritalin, Methylin) Drug tolerance, abuse, and dependence liability. Hypertension; may decrease the convulsive threshold in patients with a history of seizure disorders. Tachycardia; nervousness; insomnia; anorexia; drowsiness; dizziness.
initial: 2.5–10 mg/day
maintenance: 20–60 mg/day
dexmethylphenidate (Focalin) Dry mouth, tremor or muscle spasms, nervousness, trouble sleeping, headache, drowsiness, nausea, insomnia, increased sweating, dizziness, lightheadedness, changes in sexual function.
initial: 10 mg/day
maintenance: 10–20 mg/day

*Consult complete prescribing information for appropriate administration schedules.
Notes:
aTCAs prolong cardiac conduction through His-Purkinje system similar to Type IA antiarrhythmic agents (e.g., quinidine). They are specifically contraindicated in patients with bundle-branch disease and second- or third-degree heart block. Their effects on conduction correlate with dosage and serum concentrations and for those agents with positive chronotropic and adrenergic-stimulating properties, TCAs can cause reentry arrhythmias. Persons at greatest risk are those with preexisting cardiac conduction defects and those who have taken an overdose.
bPlasma concentrations are most useful for guiding treatment in elderly patients who are (1) experiencing signs and symptoms of toxicity, (2) unresponsive to treatment, (3) suspected of being noncompliant with planned treatment, or (4) receiving other medications that may interact or otherwise alter antidepressant medication pharmacokinetics.
cTCAs and other antidepressants may cause sexual dysfunction characterized as decreased libido, penile erectile dysfunction, and decreased sensation during orgasm and ejaculation. Management consists of waiting for spontaneous resolution with continued therapy, decreasing the antidepressant dose, selecting an alternative antidepressant, or concomitant treatment with medications that treat the dysfunction (e.g., bethanechol for antidepressants with prominent anticholinergic effects).
dCommon antimuscarinic or anticholinergic effects include dry mouth, blurred vision, constipation, and urinary retention. Although patients may eventually develop tolerance to these effects with repeated medication use, symptoms may not completely resolve until the drug is discontinued.

When selecting an antidepressant drug, it is worthwhile to consider that side effects may have a clinical advantage. For example, some TCAs, such as amitriptyline, and atypical antidepressants, such as mirtazapine and trazodone, produce sedation and may be useful for agitated patients and for those who have difficulty getting to sleep. Consequently, treatment is often initiated as a single daily dose administered at bedtime. Although most patients will develop tolerance to antidepressants' sedative effects with continued treatment, the need for soporific agents may diminish with improvement in depressive symptoms.

When selecting antidepressants, either singly or in combination, consider the following:

  • Target specific distressing symptoms.


  • Evaluate coexistent medical problems that may be exacerbated by particular antidepressants.


  • Minimize side effects and avoid worsening of current health status.


  • Determine the patient's ability to swallow solid dosage forms; he or she may be able to take an antidepressant in liquid form (e.g., amitriptyline, nortriptyline, doxepin, fluoxetine). Alternatively, some antidepressants are available as parenteral dosage forms (e.g., amitriptyline and imipramine injection).


  • Evaluate the patient's medication profile for potential interactions with antidepressant drugs.


Selective serotonin reuptake inhibitors

The postulated mechanism of action of SSRIs involves the blockade of serotonin neuronal reuptake, leading to desensitization of serotonergic feedback receptors. All currently available SSRIs are equally efficacious; they differ primarily in their safety, tolerability, half-lives, and drug-drug interactions. While some side effects are more common with some SSRIs than with others, side effects and tolerability may differ significantly in individual patients. SSRIs have become the first-line treatment for depressive disorders, owing to their better tolerability side effect profile, especially in comparison with the TCAs. As discussed earlier, antidepressant studies conducted in patients with cancer are done mostly with SSRIs or TCAs. None of the clinical trials have included or focused on children and adolescents being treated for cancer.[16-19] Overall, the evidence on the efficacy of SSRIs for treating cancer-related depression remains limited, and more studies are needed to address the efficacy, safety, tolerability, and drug-drug interaction issues in the context of cancer and cancer treatments. The British Committee on Safety of Medicines considered only one of the SSRIs (fluoxetine) to have a favorable balance of risks and benefits, but it is only considered beneficial in approximately one in ten patients.[20] Consistent with this finding, age-stratified analyses of the child and adolescent studies found that for children younger than 12 years with major depression, only fluoxetine showed benefit over placebo.[21] As noted, none of the children or adolescents in these studies had cancer, so there are no reports available that address whether there are additional increased risks of adverse events associated with the use of SSRIs following exposure to different chemotherapeutic agents and/or central nervous system (CNS) radiation treatment. Frontline, alternative, effective, behavioral, and pharmacologic treatments for depression should be used for children and adolescents being treated for cancer. However, if the risks of depression are significant and SSRIs are considered, consultation from a child psychiatrist or neurologist is essential, and close monitoring of potential adverse events is crucial. No warning has been issued for adult use of SSRIs.

Discontinuation of antidepressants

The optimal duration of antidepressant therapy for patients treated for depressive symptoms (without a depressive disorder) is unknown. Patients with a depressive disorder who achieve a beneficial response to antidepressant pharmacotherapy should continue treatment for a minimum of 4 to 6 months after depression resolves. When patients are discontinuing antidepressant medications, TCA doses should be tapered by approximately 25% per week to avoid cholinergic rebound (e.g., hypersalivation, diarrhea). In patients who experience intolerable adverse effects, however, doses may be tapered quickly. With the exception of fluoxetine, gradual tapering is advised when decreasing doses or discontinuing treatment for all SSRIs. Other antidepressants with short half-lives, such as venlafaxine, also should be tapered gradually. Withdrawal symptoms, both somatic and psychological, frequently emerge after abrupt discontinuation, during intermittent noncompliance, and sometimes during dose reduction; though these symptoms are generally mild, short-lived, and self-limiting, they can be distressing and may lead to missed workdays and decreased productivity. Mild symptoms can often be treated by reassuring a patient that they are usually transient. For more severe symptoms, it may be necessary to reinstate the dosage of the original antidepressant and slow the rate of taper. Symptoms of discontinuation may be mistaken for physical illness or relapse into depression and misdiagnosis may lead to unnecessary, costly tests and treatment. Thus, health care professionals need to be educated about the potential adverse effects of SSRI discontinuation.[22]

Side effects

TCAs can produce abnormal myocardial conduction; thus, a cardiac history and a recent EKG should be obtained in patients with a history of cardiac problems. Many tricyclic antidepressants have a sedating effect; therefore, treatment typically is started at low doses at bedtime. The main exception is desipramine, which some patients find mildly stimulating and can be administered in the morning to reduce insomnia, if it develops. Daily doses are increased slowly every few days or at weekly intervals until symptoms improve. Many patients become tolerant to the drugs' sedative effects, and total daily doses may be divided and given during patients' waking cycles.

TCAs are still regarded as first-line agents for severe, major depression; however, SSRI use is increasing for that indication because of the effectiveness of SSRIs and the low risk of clinically significant side effects that are associated with TCAs, such as cardiac arrhythmias, hypotension, and anticholinergic effects. In addition, TCAs are highly toxic on overdose. Side effects commonly associated with the SSRIs include nausea, vomiting, diarrhea, somnolence, insomnia, headache, confusion, dizziness, asthenia, and sexual dysfunction. Drug-specific adverse effects associated with fluoxetine include gastric distress, brief periods of anxiety or agitation, and anorgasmia in females. Treatment with sertraline is sometimes complicated by dyspepsia, tremor, and ejaculatory delay in men.

The pharmacokinetic profiles of SSRIs permit them to be given once a day, thus improving patient compliance.[23] Sertraline and paroxetine have a half-life of approximately 20 hours; thus, steady-state systemic concentrations can be achieved within 1 week after starting treatment and altering dosage or administration schedules. In comparison, repeated dosing appears to inhibit fluoxetine metabolism; consequently, both fluoxetine and its active metabolite, norfluoxetine, may be present in the body for weeks after discontinuing treatment.

Drug-drug interactions

Clinicians who prescribe and monitor patients receiving antidepressants should also become familiar with their potential for interactions with other medications.[24] The SSRIs venlafaxine, nefazodone, and mirtazapine are metabolized by cytochrome P450 enzymes; their pharmacokinetics may be altered, or they may affect the clearance of drugs metabolized by the same enzymes. Marked differences exist, however, between the SSRIs and SSRI metabolites with regard to their effects on specific cytochrome P450 enzymes.[15] For example, both fluoxetine and norfluoxetine inhibit CYP3A4 isoenzyme; however, the metabolite is more potent than fluoxetine and in view of its longer half-life the potential for interactions may persist for weeks after fluoxetine is discontinued.[25] Understanding the similarities and differences in their pharmacology can aid clinicians in using these agents optimally and avoiding clinically important pharmacokinetic drug-drug interactions. In addition, since all SSRIs are highly protein-bound to albumin (± alpha-1 acid glycoprotein), clinicians must consider their potential for interactions with other highly protein-bound medications. Sertraline and paroxetine may be preferred in patients with renal or hepatic dysfunction since they are metabolized and excreted as inactive compounds.[26]

Atypical antidepressants
  • Bupropion

    Bupropion is a unique alternative to tricyclics and SSRIs for treating persons with depression and cancer, especially when depression is accompanied by fatigue. Pharmacologically, bupropion is a weak inhibitor of monoamine reuptake and demonstrates a slight preference for dopamine transport inhibition; however, it may be metabolically converted to active substances with amphetamine-like activity that affect both dopamine and norepinephrine reuptake. Bupropion generally does not cause sexual dysfunction; therefore, it may be useful in treating patients who wish to remain sexually active and those who have experienced sexual dysfunction with other antidepressants. Bupropion treatment is initiated with doses of 75 mg once daily, preferably in the early part of the day. Patients may initially require a moderate- to long-acting sedative/hypnotic drug at bedtime for the insomnia, agitation, and motor restlessness sometimes associated with bupropion. Risk of seizure with bupropion may be as much as 4 times greater than is associated with other antidepressants. Single doses should not exceed 150 mg, a dose increase should not be greater than 100 mg of bupropion per day, and dose increases should be gradual—at least 3 days after a previous increase in dose. Because the risk of seizure markedly increases in patients receiving bupropion at doses between 450 mg and 650 mg, the total daily dose should not exceed 450 mg. Bupropion is contraindicated in patients with malignant diseases involving the brain and with a history of cranial trauma or seizure disorder [27] and in persons with a history of bulimia.[28]



  • Venlafaxine

    Venlafaxine affects both norepinephrine and serotonin reuptake and enhances serotonin neurotransmission.[29] Venlafaxine does not produce the same uncomfortable antimuscarinic and antiadrenergic side effects as the TCAs; however, it does produce side effects similar to the SSRIs, particularly nausea, headache, somnolence, and dry mouth. In some patients, venlafaxine may cause sustained increases in blood pressure; blood pressure should therefore be evaluated before treatment is started, monitored after treatment is initiated, and monitored after doses are increased. Venlafaxine is given twice a day, with food.



  • Trazodone and nefazodone

    The primary actions of the atypical antidepressants trazodone and nefazodone are not well established. Although they both antagonize serotonin reuptake, they are many times weaker in this respect compared with SSRIs. Trazodone is active, and both agents are metabolized to compounds that have agonistic activity at some serotonin receptors (5-HT1). Both agents may have additional active metabolites that contribute to their clinical activity.[29] Nefazodone is reported to be useful in patients with agitated depression and may be better tolerated than the SSRIs. Nefazodone can complicate some patients' management because it is a potent inhibitor of hepatic cytochrome P450 3A4 isoenzymes. Its use is, therefore, relatively contraindicated in patients who are receiving methadone and absolutely contraindicated in those receiving terfenadine or astemizole.



  • Mirtazapine

    There is growing clinical experience with mirtazapine in persons with cancer. Pharmacologically, mirtazapine is a noradrenergic and specific serotonergic antidepressant. It competitively antagonizes presynaptic alpha-adrenergic receptors (alpha2) and serotonin receptors (5-HT2 and 5-HT3), the net result of which enhances norepinephrine release and noradrenergic neurotransmission.[29,30] Sedation is the predominating side effect at subtherapeutic low doses (<15 mg/day), and anecdotal evidence suggests that sedation decreases at higher doses. Its side-effect profile also includes increased appetite, which may cause weight gain, dizziness, dry mouth, and constipation.[31] Although it is a structural analog of mianserin (an antidepressant that is marketed in Europe), mirtazapine has rarely been implicated in producing severe blood dyscrasias, including agranulocytosis, as has mianserin.[32] Little is known about mirtazapine interactions with other drugs, but it is thought to have a lesser risk of clinically significant drug interactions than SSRIs.[33] The initial dose for mirtazapine is 15 mg per day given at bedtime. Doses may be increased at intervals not less than 1 to 2 weeks, up to a maximum daily dose of 45 mg.



Benzodiazepines

Benzodiazepines can be used to effectively treat the anxiety that may be associated with depression. In patients receiving antidepressant medications and benzodiazepines concomitantly, the latter drugs may be discontinued after patients' depressive symptoms begin to abate; however, both agents can be continued safely if needed. Benzodiazepines should never be stopped abruptly because withdrawal symptoms with possible seizures may occur. The dose of benzodiazepines should be tapered slowly at a rate of approximately 25% every 3 to 4 days.

Psychostimulants

Clinical experience suggests that analeptic agents (e.g., methylphenidate and dextroamphetamine) are useful at low doses for patients whose symptoms include depressed mood, apathy, decreased energy, poor concentration, and weakness.[34] They are particularly useful for patients with advanced cancer who have a limited life expectancy (weeks to a few months). Compared with traditional antidepressants such as the TCAs and SSRIs that take 3 to 4 weeks to take effect, the psychostimulants often demonstrate antidepressant effects within a few days of starting treatment. They promote a sense of well-being, decreased fatigue, and increased appetite. Analeptic agents can be helpful in countering the sedating effects of opioids, and in comparison with antidepressants, they are rapidly effective. Adverse effects associated with analeptic agents include insomnia, euphoria, and mood lability. High doses and long-term use may produce anorexia, nightmares, insomnia, euphoria, or paranoia.

Methylphenidate and dextroamphetamine are administered in divided doses early in a patient's waking cycle to avoid sleep disturbances, e.g., insomnia and nighttime arousal. Like benzodiazepines, these medications are adjuncts to antidepressant medications; they may be started concomitant with an antidepressant and discontinued when depressive symptoms abate.[35,36]

Clinical Trials of Psychostimulants in Cancer Patients
Study  Comments  Drug(s)  Outcome 
Meyers et al. 1998 [37] Brain tumor; N = 30 methylphenidate (Ritalin) ↑ mood, ↑ cognition, ↑ function
Olin and Masand 1996 [38] Mixed cancer; N = 59; chart review dextroamphetamine (Dexedrine); methylphenidate (Ritalin) ↓ depression, ↑ appetite
Bruera et al. 1992 [39] Cancer pain vs. opioid infusion; N = 20 methylphenidate (Ritalin); placebo ↑ cognition, ↓ sedation
Fernandez et al. 1987 [36] Mixed cancer; rapid onset; N = 30 methylphenidate (Ritalin;up to 80 mg) ↓ depression
Bruera et al. 1986 [40] Pain; double-blind cross-over study; N = 24 mazindol (Mazanor) ↓ pain, ↓ appetite, no effect on mood
Joshi et al. 1982 [41] Terminally ill amphetamine ↑ comfort

Monoamine oxidase inhibitors

The use of monoamine oxidase inhibitors (MAOIs) in the cancer population has been limited because the nutritional requirements of a tyramine-free diet are generally more difficult to accomplish in patients receiving antineoplastic treatments. MAOIs are contraindicated in patients receiving opioids, sympathomimetics, and procarbazine because of the potential for developing hypertensive crisis.

MAOIs may cause adverse reactions when taken with other medications and certain foods. MAOIs impair the metabolism of morphine and other opioids as well as barbiturates and may lead to exaggerated ventilatory depression. Meperidine HCl (Demerol), an opioid, has been associated with hypertension, hyperpyrexia, skeletal muscle rigidity, seizures, and coma when used with MAOIs.[42] Exaggerated effects of antihistamines, anticholinergics, and tricyclic antidepressants may be secondary to impaired metabolism by MAOIs. In addition, the hypoglycemic effects of insulin and oral sulfonylureas may be potentiated by MAOIs.

MAOIs may also interact with specific anesthetic drugs used during surgery.[43] Cancer patients in particular may frequently undergo surgical procedures and should alert their anesthesiologist of all medications. Postoperative pain should not be treated with meperidine HCl. MAOIs should neither be taken with procarbazine, a chemotherapeutic agent used in the treatment of lymphomas and brain tumors, nor used with other antidepressants.

The U.S. Food and Drug Administration (FDA) has recently approved a transdermal antidepressant that may have particular value in the treatment of the depressed cancer patient who is unable to swallow or take medications by mouth. The antidepressant selegiline (sold under the trade name EMSAM) is an irreversible MAOI. To date, the drug has not been evaluated for the treatment of depression in cancer patients.

Many of the usual dietary restrictions (low-tyramine diet) and drug-drug interactions (the product should not be used with meperidine, propoxyphone, or methadone) are germane to selegiline (see table below). However, according to the package insert, the 20-mg skin patch (which delivers 6 mg of selegiline in a 24-hour period) can be used without the dietary restrictions found on all MAOIs marketed to date. This recommendation is supported by clinical trials and other evidence submitted to the FDA. The two higher doses (a 30-mg patch that delivers 9 mg in 24 hours and a 40-mg patch that delivers 12 mg in 24 hours) carry the usual dietary warning. This drug has not been evaluated in cancer patients for safety and efficacy.

Tyramine-Containing Foods*
Class of Food and Beverage  Tyramine-Rich Foods and Beverages To Avoid  Acceptable Foods Containing Little or No Tyramine 
Meat, poultry, and fish Air-dried, aged, and fermented meats, sausages, and salamis (including cacciatore, hard salami, and mortadella); pickled herring; and any spoiled or improperly stored meat, poultry, and fish (e.g., foods that have undergone changes in color or odor or that have become moldy); spoiled or improperly stored animal livers Fresh meat, poultry, and fish, including fresh processed meats (e.g., lunch meats, hot dogs, breakfast sausage, and cooked sliced ham)
Vegetables Broad bean pods (fava bean pods) All other vegetables
Dairy Aged cheeses Processed cheeses, mozzarella, ricotta cheese, cottage cheese, and yogurt
Beverages All varieties of tap beer, and beers that have not been pasteurized so as to allow for ongoing fermentation As with other antidepressants, concomitant use of alcohol with selegiline is not recommended. (Bottled and canned beers and wines contain little or no tyramine.)
Miscellaneous Concentrated yeast extract (e.g., Marmite), sauerkraut, most soybean products (including soy sauce and tofu); OTC supplements containing tyramine Brewer’s yeast, baker’s yeast, soy milk, commercial chain-restaurant pizzas prepared with cheeses low in tyramine

OTC = over-the-counter.
*Adapted from the EMSAM Medication Guide.[44] The foods and beverages listed above should be avoided beginning on the first day of treatment with selegiline 9 mg/24 h or 12 mg/24 h and should continue to be avoided for 2 weeks after a dose reduction to 6 mg/24 h or following the discontinuation of selegiline 9 mg/24 h or 12 mg/24 h.

Selegiline is a nonselective MAOI, inhibiting not only the MAO-B enzyme in the central nervous system but also MAO-A elsewhere in the body. In the digestive tract, MAO-A normally metabolizes tyramine, a dietary amine that is found in high concentrations in foods such as aged cheese and red wine. The breakdown of tyramine in the gut prevents significant amounts of it from being absorbed and circulated throughout the body. Tyramine is a potent pressor—leading to constriction of blood vessels—which ultimately results in increased blood pressure. Large amounts of tyramine can lead to hypertensive crises, resulting in stroke, heart attack, and even death. Because the medication is absorbed from the skin patch and bypasses the gut wall, it is thought that transdermal selegiline will have a significantly reduced effect on MAO-A in the digestive tract. In addition, at lower doses, selegiline is thought to inhibit MAO-B preferentially, while at higher doses both A and B isoenzymes are affected. With significantly reduced inhibition of digestive tract MAO-A, dietary restrictions are not considered necessary for the lower dose. In considering starting this drug, consult with a pharmacist about multiple classes of drug-drug interactions. This drug has not been evaluated in people with cancer.[45]

Foods that contain large amounts of tyramine, such as cheese, chicken liver, chocolate, beer, and wine, may provoke hypertension (initially manifesting as headache) and cardiac dysrhythmias.

St. John's wort

There has been much interest in the use of St. John's wort (SJW, Hypericum perforatum) as an herbal antidepressant. Its use has become widely advertised as an over-the-counter supplement for mood enhancement.

In the United States, dietary supplements are regulated as foods, not drugs. Premarket approval by the FDA is not required unless specific disease prevention or treatment claims are made, which is often not the case with SJW. Because a review for manufacturing consistency is not required for dietary supplements and no specific standards for dose or purity exist, there may be considerable variation from lot to lot for all products marketed as dietary supplements, including SJW.

Promotional statements about SJW may address mood enhancement and positive outlook. It may be found as a stand-alone supplement or in combination with other herbs, vitamin and mineral supplements, or food products such as teas. The FDA issued a warning highlighting the results from a study conducted by the National Institutes of Health that showed a significant drug interaction between SJW and indinavir, a protease inhibitor used to treat HIV infection. In this study, concomitant administration of SJW and indinavir substantially decreased indinavir plasma concentrations, potentially due to induction of the cytochrome P450 metabolic pathway.[46]

While its specific mechanisms of action are unclear, there are 3 presumed active compounds in the herb. One of these compounds is a mild MAOI (see above precautions). Historically, prescription MAOIs have been used to treat depression as well as Parkinson disease, narcolepsy, and occasionally hypertension, but they are rarely used today. The side effects of SJW include dry mouth, dizziness, gastrointestinal (GI) distress, fatigue, and confusion.

A randomized double-blind placebo-controlled trial involving 200 adult outpatients with major depressive disorder compared the safety and efficacy of SJW with placebo. This study was designed to address the numerous methodological flaws in the existing literature [47] that served as the basis for the meta-analysis that had concluded that SJW is significantly superior to placebo.[48] This study represents the first report of a large-scale randomized placebo-controlled trial of SJW in the United States. This study does not support significant antidepressant or antianxiety effects of SJW. Specifically, compared with placebo, there were no significant differences in response rates in the overall sample of outpatients with major depression. On the basis of these trial data indicating lack of efficacy in depressed patients in primary care settings, as well as the lack of properly designed positive trials in cancer patients, SJW should not be recommended for major depression in cancer patients.

Antidepressant effects

The following tables highlight tips that may be useful in determining what medication is best to use for a particular patient. The tables focus on the effects these medications may have beyond their antidepressant effects that may decrease or increase patient distress, such as fatigue, insomnia, and nausea and vomiting.

Physical Symptom- and Distress-Driven Approach to Choosing an Antidepressant in Adult Cancer Patients
Distressing Symptom   SSRI  TCA   Psychostimulants   Other 
Fatigue + (a) + + (a)
Insomnia (b) + + (b)
Neuropathic pain (c) + +
Opioid side effects + +
Constipation + +
Loss of appetite (weight loss) + +
Anxiety + + + (d)
Dry mouth/stomatitis + - +

Key:
(-) use of this medication could worsen the symptom
(+) use of this medication could relieve the symptom
Notes:
(a) Although all SSRIs have the potential paradoxical side effect of hypersomnia, fluoxetine is particularly activating. Bupropion is also somewhat activating.
(b) Sedating antidepressants are useful for insomnia, either alone or in addition to another antidepressant. Trazodone and mirtazapine are often used as sleep aids in combination with another antidepressant.
(c) Some antidepressants are useful in treating neuropathic pain. The most studied of these are the TCAs, particularly amitriptyline.
(d) Sedating antidepressants are most useful for anxious/agitated patients. These include the TCAs, trazodone, mirtazapine, and nefazodone.
SSRI = Selective Serotonin Reuptake Inhibitor
TCA = Tricyclic Antidepressant
In general, doses should start low and increase slowly. This list does not indicate absolute indications or contraindications for particular medications. A current Physicians' Desk Reference or another reliable drug information resource and experience should guide clinical decision making.

Factors to Consider in Choosing an Antidepressant For Adult Cancer Patients
Comorbid Medical Conditions  SSRI  TCA   Psychostimulants   Other 
Cardiac history + - + (a)
Hepatic dysfunction + (b) + -
Renal dysfunction (c)
Glaucoma + - (d)
Neuropathic pain + +

Key:
(-) use of this medication may be a less appropriate choice
(+) use of this medication could relieve the symptom
Notes:
(a) In general, TCAs and psychostimulants can cause and exacerbate cardiac arrhythmia. SSRIs, bupropion, venlafaxine, and nefazodone are generally less likely to cause cardiac problems. EKGs should be obtained before starting TCA medication, and a cardiologist should be consulted if there is concern for cardiac compromise.
(b) The shorter-acting SSRIs (sertraline and paroxetine) are less problematic than fluoxetine in patients with hepatic dysfunction. There is less potential for adverse drug interactions and fewer problems related to drug accumulation due to a shorter half-life. Sertraline and nefazodone reportedly have less effect on hepatic P450 enzyme activity.
(c) Clinicians should consider whether antidepressant doses and administration schedules require modification for their patients with renal or hepatic insufficiency.
(d) The TCAs are contraindicated in closed-angle glaucoma.

It should be noted that electroconvulsive therapy (ECT) is a useful and safe therapy when other interventions have not succeeded in relieving the depressive syndrome that may represent a life-threatening complication of treatable cancer.[49,50] Experience is limited, however, in using ECT in patients receiving mirtazapine and trazodone, and there are no clinical studies establishing the use of ECT in patients receiving SSRIs. Prolonged seizures have occurred rarely in patients receiving fluoxetine.

Psychotherapy

Overview

Traditionally, depressive symptomatology was managed with insight-oriented psychotherapy, which is quite useful for some people with cancer. For many other people, these symptoms are best managed with some combination of crisis intervention, brief supportive psychotherapy, and cognitive-behavioral techniques.

Psychotherapy for depression has been offered in a variety of forms. Most interventions have been time limited (ranging between 4 and 30 hours), have been offered in both individual and small-group formats, and have included a structured educational component about cancer or a specific relaxation component.[51]

Cognitive-behavioral psychotherapy has been one of the most prominent types of therapies studied in recent investigations. Cognitive-behavioral interventions focus on altering specific coping strategies aimed at improving overall adjustment and typically focus on specific thoughts and their relationship to emotions and behaviors. Understanding and altering one’s thoughts can change emotional reactions and accompanying behaviors. For example, frequent, intrusive, uncontrollable thoughts about loss, life changes, or death can cause poor concentration and precipitate feelings of sadness, guilt, and worthlessness. In turn, these feelings can result in increased sleep, withdrawal, and isolation. A cognitive-behavioral intervention focuses on the intrusive thoughts, often challenging their accuracy or rationality and noting specific patterns of cognitive distortions. Simultaneously, patients develop specific cognitive coping strategies that are designed to alter emotional reactions and accompanying behaviors. The end result is improved coping, enhanced adjustment, and better overall quality of life.

Other goals of psychotherapy include enhancing coping skills, directly reducing distress, improving problem-solving skills, mobilizing support, reshaping negative or self-defeating thoughts, and developing a close personal bond with a knowledgeable, empathic health care provider.[52-56] Consultation with a cleric or a member of a pastoral care department may also help some individuals.

Specific goals of these therapies include the following:

  • Assist people with cancer and their families by answering questions about the illness and its treatment, clarifying information, correcting misunderstandings, giving reassurance, and normalizing responses to the illness and its effect on their families. Explore the present situation with the patient and how it relates to his or her previous experiences with cancer.


  • Assist with problem solving, bolster the patient’s usual adaptive defenses, and help the patient and family develop further supportive and adaptive coping mechanisms. Identify maladaptive coping mechanisms and assist the family in developing alternative coping strategies. Explore areas of related stressors (e.g., family role and lifestyle changes), and encourage family members to support and share concerns with each other.


  • When the focus of treatment changes from cure to palliation, reinforce strongly that, though curative treatment has ended, the team will aggressively treat symptoms as part of the palliation plan; the patient and family will not be abandoned; and staff members will work very hard to maintain comfort, control pain, and maintain the dignity of the patient and his or her family members.


Cancer support groups can be useful adjunctive therapies in the treatment of cancer patients.[57,58] Recent support group interventions have demonstrated significant effects on mood disturbance, use of positive coping strategies, improvement in quality of life, and positive immune responses.[59-61] Support groups can be found through The Wellness Community, the American Cancer Society, and many other community resources, including the social work departments of medical centers or hospitals.

Empirical studies of the efficacy of psychotherapy

Psychotherapy as a treatment for depression in the general adult mental health population has been extensively researched and found to be effective.[62] Recent reviews have also concluded that psychotherapy is an effective intervention for cancer patients experiencing depression.[63,51] In studies designed to prevent the occurrence of depression (i.e., patients not selected because of their depressive symptoms), intervention effects are positive, though small to moderate effect sizes have been reported (effect sizes range from 0.19 to 0.54).[51] However, in those studies in which patients were intentionally selected because they exhibited depressive symptoms, intervention effects were strong (effect size, 0.94).[63] An effect size of 0.94 indicates that the average patient in the treatment group was advantaged, compared with approximately 82% of patients in the control group.

One well-designed randomized clinical trial of a cognitive-behavioral intervention for depressed cancer patients investigated the effect of training in problem solving on symptoms of depression.[64] The intervention consisted of 10 1.5-hour weekly individual psychotherapy sessions focused on training to become an effective problem solver. Problem-solving tasks were emphasized, including skills in (a) better defining and formulating the nature of problems, (b) generating a wide range of alternative solutions, (c) systematically evaluating consequences of a solution while deciding on an optimal one, and (d) evaluating outcome after solution implementation. Between-session homework with tasks relevant to each step was assigned, and patients were provided with a written manual and encouraged to refer to it as problems arose. One hundred thirty-two adult cancer patients were randomly assigned to the problem-solving treatment or a wait-list control. Overall results showed both improved problem-solving abilities and clinically significant decreases in symptoms of depression.

References

  1. Whooley MA, Simon GE: Managing depression in medical outpatients. N Engl J Med 343 (26): 1942-50, 2000.  [PUBMED Abstract]

  2. Derogatis LR, Morrow GR, Fetting J, et al.: The prevalence of psychiatric disorders among cancer patients. JAMA 249 (6): 751-7, 1983.  [PUBMED Abstract]

  3. Endicott J: Measurement of depression in patients with cancer. Cancer 53 (10 Suppl): 2243-9, 1984.  [PUBMED Abstract]

  4. Massie MJ, Holland JC: Consultation and liaison issues in cancer care. Psychiatr Med 5 (4): 343-59, 1987.  [PUBMED Abstract]

  5. Fisch M: Treatment of depression in cancer. J Natl Cancer Inst Monogr (32): 105-11, 2004.  [PUBMED Abstract]

  6. Ashbury FD, Madlensky L, Raich P, et al.: Antidepressant prescribing in community cancer care. Support Care Cancer 11 (5): 278-85, 2003.  [PUBMED Abstract]

  7. Williams JW Jr, Mulrow CD, Chiquette E, et al.: A systematic review of newer pharmacotherapies for depression in adults: evidence report summary. Ann Intern Med 132 (9): 743-56, 2000.  [PUBMED Abstract]

  8. Fisch MJ, Callahan CM, Kesterson JG, et al.: The use of an electronic patient record system to identify advanced cancer patients and antidepressant drug use. J Palliat Med 2 (4): 403-9, 1999. 

  9. Holland JC, Romano SJ, Heiligenstein JH, et al.: A controlled trial of fluoxetine and desipramine in depressed women with advanced cancer. Psychooncology 7 (4): 291-300, 1998 Jul-Aug.  [PUBMED Abstract]

  10. Pezzella G, Moslinger-Gehmayr R, Contu A: Treatment of depression in patients with breast cancer: a comparison between paroxetine and amitriptyline. Breast Cancer Res Treat 70 (1): 1-10, 2001.  [PUBMED Abstract]

  11. Fisch MJ, Loehrer PJ, Kristeller J, et al.: Fluoxetine versus placebo in advanced cancer outpatients: a double-blinded trial of the Hoosier Oncology Group. J Clin Oncol 21 (10): 1937-43, 2003.  [PUBMED Abstract]

  12. Safety Review of Antidepressants Used by Children Completed. London, UK: Medicines and Healthcare Products Regulatory Agency, 2003. Available online. Last accessed December 15, 2008. 

  13. U.S. Food and Drug Administration.: Antidepressant Use in Children, Adolescents, and Adults. Rockville, Md: Food and Drug Administration, Center for Drug Evaluation and Research, 2007. Available online. Last accessed December 15, 2008. 

  14. Musselman DL, Lawson DH, Gumnick JF, et al.: Paroxetine for the prevention of depression induced by high-dose interferon alfa. N Engl J Med 344 (13): 961-6, 2001.  [PUBMED Abstract]

  15. Preskorn SH: Clinically relevant pharmacology of selective serotonin reuptake inhibitors. An overview with emphasis on pharmacokinetics and effects on oxidative drug metabolism. Clin Pharmacokinet 32 (Suppl 1): 1-21, 1997.  [PUBMED Abstract]

  16. Emslie GJ, Rush AJ, Weinberg WA, et al.: A double-blind, randomized, placebo-controlled trial of fluoxetine in children and adolescents with depression. Arch Gen Psychiatry 54 (11): 1031-7, 1997.  [PUBMED Abstract]

  17. Emslie GJ, Heiligenstein JH, Wagner KD, et al.: Fluoxetine for acute treatment of depression in children and adolescents: a placebo-controlled, randomized clinical trial. J Am Acad Child Adolesc Psychiatry 41 (10): 1205-15, 2002.  [PUBMED Abstract]

  18. Keller MB, Ryan ND, Strober M, et al.: Efficacy of paroxetine in the treatment of adolescent major depression: a randomized, controlled trial. J Am Acad Child Adolesc Psychiatry 40 (7): 762-72, 2001.  [PUBMED Abstract]

  19. Wagner KD, Ambrosini P, Rynn M, et al.: Efficacy of sertraline in the treatment of children and adolescents with major depressive disorder: two randomized controlled trials. JAMA 290 (8): 1033-41, 2003.  [PUBMED Abstract]

  20. Ramchandani P: Treatment of major depressive disorder in children and adolescents. BMJ 328 (7430): 3-4, 2004.  [PUBMED Abstract]

  21. Bridge JA, Iyengar S, Salary CB, et al.: Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA 297 (15): 1683-96, 2007.  [PUBMED Abstract]

  22. Rosenbaum JF, Zajecka J: Clinical management of antidepressant discontinuation. J Clin Psychiatry 58 (Suppl 7): 37-40, 1997.  [PUBMED Abstract]

  23. Bruera E, Brenneis C, Paterson AH, et al.: Use of methylphenidate as an adjuvant to narcotic analgesics in patients with advanced cancer. J Pain Symptom Manage 4 (1): 3-6, 1989.  [PUBMED Abstract]

  24. Richelson E: Pharmacokinetic drug interactions of new antidepressants: a review of the effects on the metabolism of other drugs. Mayo Clin Proc 72 (9): 835-47, 1997.  [PUBMED Abstract]

  25. Caccia S: Metabolism of the newer antidepressants. An overview of the pharmacological and pharmacokinetic implications. Clin Pharmacokinet 34 (4): 281-302, 1998.  [PUBMED Abstract]

  26. Jin Y, Desta Z, Stearns V, et al.: CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst 97 (1): 30-9, 2005.  [PUBMED Abstract]

  27. Physician's Desk Reference. 51st ed. Montvale, NJ: Medical Economics, 1997. 

  28. Horne RL, Ferguson JM, Pope HG Jr, et al.: Treatment of bulimia with bupropion: a multicenter controlled trial. J Clin Psychiatry 49 (7): 262-6, 1988.  [PUBMED Abstract]

  29. Stahl SM: Basic psychopharmacology of antidepressants, part 1: Antidepressants have seven distinct mechanisms of action. J Clin Psychiatry 59 (Suppl 4): 5-14, 1998.  [PUBMED Abstract]

  30. Theobald DE, Kirsh KL, Holtsclaw E, et al.: An open-label, crossover trial of mirtazapine (15 and 30 mg) in cancer patients with pain and other distressing symptoms. J Pain Symptom Manage 23 (5): 442-7, 2002.  [PUBMED Abstract]

  31. Montgomery SA: Safety of mirtazapine: a review. Int Clin Psychopharmacol 10 (Suppl 4): 37-45, 1995.  [PUBMED Abstract]

  32. Chaplin S: Bone marrow depression due to mianserin, phenylbutazone, oxyphenbutazone, and chloramphenicol--Part II. Adverse Drug React Acute Poisoning Rev 5 (3): 181-96, 1986 Autumn.  [PUBMED Abstract]

  33. Owen JR, Nemeroff CB: New antidepressants and the cytochrome P450 system: focus on venlafaxine, nefazodone, and mirtazapine. Depress Anxiety 7 (Suppl 1): 24-32, 1998.  [PUBMED Abstract]

  34. Homsi J, Nelson KA, Sarhill N, et al.: A phase II study of methylphenidate for depression in advanced cancer. Am J Hosp Palliat Care 18 (6): 403-7, 2001 Nov-Dec.  [PUBMED Abstract]

  35. Feighner JP, Boyer WF: Perspectives in Psychiatry. Volume 1. Selective Serotonin Re-uptake Inhibitors: The Clinical Use of Citalopram, Fluoxetine, Fluvoxamine, Paroxetine, and Sertraline. New York, NY: John Wiley & Sons Ltd, 1991. 

  36. Fernandez F, Adams F, Holmes VF, et al.: Methylphenidate for depressive disorders in cancer patients. An alternative to standard antidepressants. Psychosomatics 28 (9): 455-61, 1987.  [PUBMED Abstract]

  37. Meyers CA, Weitzner MA, Valentine AD, et al.: Methylphenidate therapy improves cognition, mood, and function of brain tumor patients. J Clin Oncol 16 (7): 2522-7, 1998.  [PUBMED Abstract]

  38. Olin J, Masand P: Psychostimulants for depression in hospitalized cancer patients. Psychosomatics 37 (1): 57-62, 1996 Jan-Feb.  [PUBMED Abstract]

  39. Bruera E, Miller MJ, Macmillan K, et al.: Neuropsychological effects of methylphenidate in patients receiving a continuous infusion of narcotics for cancer pain. Pain 48 (2): 163-6, 1992.  [PUBMED Abstract]

  40. Bruera E, Carraro S, Roca E, et al.: Double-blind evaluation of the effects of mazindol on pain, depression, anxiety, appetite, and activity in terminal cancer patients. Cancer Treat Rep 70 (2): 295-8, 1986.  [PUBMED Abstract]

  41. Joshi JH, de Jongh CA, Schnaper N, et al.: Amphetamine therapy for enhancing the comfort of terminally ill patients with cancer. Proceedings of the American Society of Clinical Oncology 1: 55, 1982. 

  42. Brown TC, Cass NM: Beware -- the use of MAO inhibitors is increasing again. Anaesth Intensive Care 7 (1): 65-8, 1979.  [PUBMED Abstract]

  43. Monoamine oxidase inhibitors. In: Stoelting RK, Dierdorf SF, McCammon RL, eds.: Anesthesia and Co-Existing Disease. 2nd ed. New York, NY: Churchill Livingstone, 1988, pp 720-2. 

  44. Medication Guide: EMSAM. Princeton, NJ: Bristol-Myers Squibb Company, 2006. Available online. Last accessed December 15, 2008. 

  45. EMSAM (selegiline transdermal system): continuous delivery for once-daily application. Princeton, NJ: Bristol-Myers Squibb Company, 2006. Available online. Last accessed December 15, 2008. 

  46. FDA Public Health Advisory (February 10, 2000): Risk of Drug Interactions With St John's Wort and Indinavir and Other Drugs. Rockville, Md: Food and Drug Administration, Center for Drug Evaluation and Research, 2001. Available online. Last accessed December 15, 2008. 

  47. Shelton RC, Keller MB, Gelenberg A, et al.: Effectiveness of St John's wort in major depression: a randomized controlled trial. JAMA 285 (15): 1978-86, 2001.  [PUBMED Abstract]

  48. Linde K, Mulrow CD: St John's wort for depression. Cochrane Database Syst Rev (2): CD000448, 2000.  [PUBMED Abstract]

  49. Massie MJ, Lesko LM: Psychopharmacological management. In: Holland JC, Rowland JH, eds.: Handbook of Psychooncology: Psychological Care of the Patient With Cancer. New York, NY: Oxford University Press, 1989, pp 470-91. 

  50. Massie MJ, Shakin EJ: Management of depression and anxiety in cancer patients. In: Breitbart W, Holland JC, eds.: Psychiatric Aspects of Symptom Management in Cancer Patients. Washington, DC: American Psychiatric Press, 1993, pp 470-91. 

  51. Barsevick AM, Sweeney C, Haney E, et al.: A systematic qualitative analysis of psychoeducational interventions for depression in patients with cancer. Oncol Nurs Forum 29 (1): 73-84; quiz 85-7, 2002 Jan-Feb.  [PUBMED Abstract]

  52. Forester B, Kornfeld DS, Fleiss JL: Psychotherapy during radiotherapy: effects on emotional and physical distress. Am J Psychiatry 142 (1): 22-7, 1985.  [PUBMED Abstract]

  53. Holland JC, Morrow GR, Schmale A, et al.: A randomized clinical trial of alprazolam versus progressive muscle relaxation in cancer patients with anxiety and depressive symptoms. J Clin Oncol 9 (6): 1004-11, 1991.  [PUBMED Abstract]

  54. Worden JW, Weisman AD: Preventive psychosocial intervention with newly diagnosed cancer patients. Gen Hosp Psychiatry 6 (4): 243-9, 1984.  [PUBMED Abstract]

  55. Lovejoy NC, Matteis M: Cognitive-behavioral interventions to manage depression in patients with cancer: research and theoretical initiatives. Cancer Nurs 20 (3): 155-67, 1997.  [PUBMED Abstract]

  56. Greer S, Moorey S, Baruch JD, et al.: Adjuvant psychological therapy for patients with cancer: a prospective randomised trial. BMJ 304 (6828): 675-80, 1992.  [PUBMED Abstract]

  57. Cain EN, Kohorn EI, Quinlan DM, et al.: Psychosocial benefits of a cancer support group. Cancer 57 (1): 183-9, 1986.  [PUBMED Abstract]

  58. Montazeri A, Jarvandi S, Haghighat S, et al.: Anxiety and depression in breast cancer patients before and after participation in a cancer support group. Patient Educ Couns 45 (3): 195-8, 2001.  [PUBMED Abstract]

  59. Fawzy FI, Fawzy NW, Hyun CS, et al.: Malignant melanoma. Effects of an early structured psychiatric intervention, coping, and affective state on recurrence and survival 6 years later. Arch Gen Psychiatry 50 (9): 681-9, 1993.  [PUBMED Abstract]

  60. Spiegel D, Glafkides MC: Effects of group confrontation with death and dying. Int J Group Psychother 33 (4): 433-47, 1983.  [PUBMED Abstract]

  61. Spiegel D, Bloom JR, Yalom I: Group support for patients with metastatic cancer. A randomized outcome study. Arch Gen Psychiatry 38 (5): 527-33, 1981.  [PUBMED Abstract]

  62. Lambert MJ, Ogles BM: The efficacy and effectiveness of psychotherapy. In: Lambert MJ: Bergin and Garfield's Handbook of Psychotherapy and Behavior Change. 5th ed. New York: John Wiley & Sons Inc, 2004, pp 139-93. 

  63. Sheard T, Maguire P: The effect of psychological interventions on anxiety and depression in cancer patients: results of two meta-analyses. Br J Cancer 80 (11): 1770-80, 1999.  [PUBMED Abstract]

  64. Nezu AM, Nezu CM, Felgoise SH, et al.: Project Genesis: assessing the efficacy of problem-solving therapy for distressed adult cancer patients. J Consult Clin Psychol 71 (6): 1036-48, 2003.  [PUBMED Abstract]

Back to Top

< Previous Section  |  Next Section >


A Service of the National Cancer Institute
Department of Health and Human Services National Institutes of Health USA.gov